US20190300957A1 - Methods of diagnosing inflammatory bowel disease through rnaset2 - Google Patents

Methods of diagnosing inflammatory bowel disease through rnaset2 Download PDF

Info

Publication number
US20190300957A1
US20190300957A1 US16/084,858 US201716084858A US2019300957A1 US 20190300957 A1 US20190300957 A1 US 20190300957A1 US 201716084858 A US201716084858 A US 201716084858A US 2019300957 A1 US2019300957 A1 US 2019300957A1
Authority
US
United States
Prior art keywords
rnaset2
disease
subject
ibd
tl1a
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US16/084,858
Other versions
US11186872B2 (en
Inventor
Rebecca Gonsky
Stephan R. Targan
Richard L. DEEM
Philip Fleshner
Dermot P. McGovern
Janine Bilsborough
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cedars Sinai Medical Center
Original Assignee
Cedars Sinai Medical Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cedars Sinai Medical Center filed Critical Cedars Sinai Medical Center
Priority to US16/084,858 priority Critical patent/US11186872B2/en
Assigned to CEDARS-SINAI MEDICAL CENTER reassignment CEDARS-SINAI MEDICAL CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BILSBOROUGH, JANINE, DEEM, Richard L., FLESHNER, Philip, GONSKY, Rebecca, MCGOVERN, DERMOT P., TARGAN, STEPHEN R.
Publication of US20190300957A1 publication Critical patent/US20190300957A1/en
Application granted granted Critical
Publication of US11186872B2 publication Critical patent/US11186872B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/154Methylation markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/065Bowel diseases, e.g. Crohn, ulcerative colitis, IBS

Definitions

  • the invention relates to inflammatory bowel disease and RNASET2 as a biomarker for disease severity and targeting anti-TL1A therapy.
  • IBD Inflammatory bowel disease
  • CD Crohn's disease
  • UC ulcerative colitis
  • IBD is generally believed to be triggered in genetically susceptible individuals by an inappropriate immune response to the commensal flora.
  • the high clinical heterogeneity and genetic complexity of CD and UC suggest that the underlying biological pathways driving disease almost certainly differ in subgroups of patients.
  • the development of early and targeted therapeutics requires subgroup stratification and prognostic biomarker identification, particularly in predicting an overall mild, compared to severe, disease course.
  • IBD susceptibility loci have been identified, little is known regarding their functional significance. Genetic variation in TNFSF15 is associated with CD in multiple populations, and the protein it encodes, TL1A, is a key mediator of mucosal inflammation.
  • TL1A expression is up-regulated in inflamed regions of the intestine in both CD and UC.
  • IBD patients elevated TL1A levels correlate with TNFSF15 genotype and disease severity.
  • CD patients with elevated serum/tissue levels of TL1A have increased risk of developing fibrosis/stricturing disease behavior.
  • TL1A synergizes with interleukin 12 (IL-12) and interleukin 18 (IL-18) (12/18), leading to rapid enhancement of IFN- ⁇ production, another key mediator of mucosal inflammation.
  • IL-12 interleukin 12
  • IL-18 interleukin 18
  • FIG. 1 depicts RNASET2 eQTL microarray in uninvolved small intestine, in accordance with various embodiments of the invention.
  • FIG. 2 shows that the RNASET2 major allele is associated with decreased expression of RNASET2 in sigmoid colon and rectum of CD patients, in accordance with various embodiments of the invention.
  • FIG. 3 shows that the RNASET2 major allele is associated with decreased expression of RNASET2 in sigmoid colon and rectum of UC patients, in accordance with various embodiments of the invention.
  • FIG. 4 shows that the RNASET2 risk allele is associated with decreased expression of RNASET2 in inflamed large bowel in CD and UC patients, in accordance with various embodiments of the invention. Similar results were observed for rs1819333.
  • FIG. 5 shows that the RNASET2 major allele is associated with decreased RNASET2 expression in small bowel from CD surgeries, in accordance with various embodiments of the invention.
  • FIG. 6 depicts RNASET2 eQTL in EBV Transformed B Cell Lines, in accordance with various embodiments of the invention.
  • the major allele is associated with lower levels of RNASET2 mRNA expression in EBV transformed B cell lines.
  • FIG. 7 depicts RNASET2 expression following IL-12, IL-18, and/or TL1A and treatment, in accordance with various embodiments of the invention.
  • FIGS. 8A-8C depict RNASET2 expression in CD patients, in accordance with various embodiments of the invention.
  • FIG. 9 shows a decreased expression of RNASET2 in IBD patients with RNASET2 risk alleles, in accordance with various embodiments of the invention.
  • FIG. 10 depicts RNASET2 methylation versus GWAS p values in CD and UC patients, in accordance with various embodiments of the invention.
  • FIG. 11 depicts the eQTL of RNASET2 in refractory IBD, in accordance with various embodiments of the invention.
  • RNASET2 SNPs rs2149085, rs1819333 and rs9355610
  • CD3+ peripheral T cells from 11 CD and 10 UC patients requiring surgical intervention for disease management, using an Illumina expression array.
  • FIG. 12 depicts the eQTL of RNASET2 in CD small bowel (ileal) surgical resection of 85 CD patients using an Agilent expression array, in accordance with various embodiments of the invention.
  • FIGS. 13A-13D depict the mQTL of RNASET2 in refractory IBD, in accordance with various embodiments of the invention.
  • FIGS. 14A-14B depict the mapping of eQTL and mQTL across RNASET2 in patients with refractory or mild disease, in accordance with various embodiments of the invention.
  • FIG. 15 depicts sorting and IFN ⁇ expression of CD4+ T cells stimulated with IL-12, IL-18 and/or TL1A, in accordance with various embodiments of the invention. Histograms of side scatter vs. IFN- ⁇ for CD4+T cells stimulated with recombinant human IL-12 (500 pg/ml) and IL-18 (50 ng/ml) and TL1A (100 ng/ml) for 8 h from 4 donor (D1-4).
  • FIG. 16 depicts a dendrogram of hierarchical clustering using centered correlation and average linkage, in accordance with various embodiments of the invention.
  • FIG. 17 depicts a class prediction analysis classifying the IFN ⁇ -secreting and non-secreting subgroups based on expression level, in accordance with various embodiments of the invention. Heatmap of 764 predictor genes.
  • FIG. 18 depicts a proportion of genes differentially expressed that was increased in GWAS versus other regions, in accordance with various embodiments of the invention.
  • FIG. 19 depicts 183 transcribed IBD associated SNP regions in the T cells, in accordance with various embodiments of the invention.
  • FIG. 20 depicts a volcano plot of the class predictor GWAS transcripts of IBD risk predictor genes, in accordance with various embodiments of the invention.
  • FIGS. 21A-21B show that silencing RNASET2 enhances IFN- ⁇ secretion, in accordance with various embodiments of the invention.
  • FIG. 22 demonstrates the inverse correlation of IFN- ⁇ and RNASET2 expression in accordance with various embodiments of the invention.
  • FIG. 23 demonstrates a negative correlation of RNASET2 methylation and expression in CD3 + T cells (cg25258033, located 1.4 kb within the first intron) in 21 IBD patients, in accordance with various embodiments of the invention.
  • FIGS. 24A-24D depict correlation of RNASET2 and TNFSF15 expression in CD3+ T cells from patients with refractory disease requiring surgical intervention for disease management, using RNA-seq, in accordance with various embodiments of the invention.
  • FIG. 25 depicts the correlation expression of RNASET2 versus A) PU.1 and B) ELF1, in accordance with various embodiments of the invention.
  • the risk SNP rs2149092 C/T abolishes the IRF4, PU.1, and ELF-1 binding site.
  • C—non-risk and T risk allele.
  • FIG. 26 depicts correlation of expression and methylation located within 100 kb of the RNASET2 transcriptional start site in 21 IBD patients, in accordance with various embodiments of the invention.
  • FIG. 27 depicts IFN- ⁇ expression of IFN- ⁇ producing and non-producing levels in CD4+ T cells, in accordance with various embodiments of the invention.
  • FIG. 28 depicts the correlation of GWAS p values with eQTL p values over the RNASET2 locus, in accordance with various embodiments of the invention.
  • GWAS p values are based upon data from 18729 CD and 34897 controls.
  • eQTL p values are based upon genotyping and RNA-seq based expression of RNASET2 for 71 CD patients with refractory disease, requiring surgical intervention for disease management.
  • FIG. 29 depicts the effect of RNASET2 silencing on IFN- ⁇ secretion, in accordance with various embodiments of the invention. Inhibition of RNASET2 expression by RNASET2-specific siRNA was greater than 50% in all experiments.
  • FIG. 30 depicts an association of decreased expression of RNASET2 with ASCA Sero-positivity, in accordance with various embodiments of the invention.
  • FIG. 31 depicts an association of a decreased expression of RNASET2 with penetrating disease, in accordance with various embodiments of the invention.
  • FIG. 32 depicts that patients with RNASET2 disease associated SNPs exhibited a shorter time to reoperation, in accordance with various embodiments of the invention. Time between surgeries based upon carriage for IBD risk SNP rs9355610 (SEQ ID NO: 3) for 154 CD patients who underwent multiple surgeries.
  • FIG. 33 depicts that the rs2149092 SNP (SEQ ID NO: 2) Alters DNA Shape.
  • FIG. 34 depicts the correlation of RNASET2 versus Ets1 expression, in accordance with various embodiments of the invention.
  • FIG. 35 depicts that the rs2149092 SNP (SEQ ID NO: 2) distorts DNA shape at the Ets1 binding site.
  • FIGS. 36A-36C depict identification of potential regulatory function of RNASET2 disease associated variant rs2149092 (SEQ ID NO: 2) the prospective regulatory role of RNASET2 variant rs2149092 (C-non-risk allele/T-risk allele), in accordance with various embodiments of the invention.
  • FIGS. 37A-37G depict the effect of RNASET2 silencing on IFN- ⁇ secretion and cellular aggregation, in accordance with various embodiments of the invention.
  • FIG. 38 depicts in accordance with various embodiments of the invention, the association of RNASET2 disease risk variant SNPs at the time of surgery with therapeutic failure of thiopurine or anti-TNF therapy, ANCA sero-positivity and an increased length of intestinal resection (data summarized in Table 18).
  • FIG. 39 depicts in accordance with various embodiments of the invention, the association of RNASET2 disease risk variant SNPs with disease recurrence in 38 patients who were not receiving postoperative prophylaxis. Post-operative endoscopies were performed and classified by Rutgeerts score. (data summarized in Table 18).
  • FIG. 40 depicts tissue-specific functional annotation of RNASET2 locus. Heatmap of H3K4me3, H3K4me1 and RNAseq data from REMC.
  • FIG. 41 depicts the correlation of RNASET2 expression and multiple ETS and IRF4 transcription factors in CD3+peripheral T cells from 108 CD patients with refractory disease, using RNA-seq.
  • FIG. 42 depicts in accordance with various embodiments of the invention, a heat-map illustrating protein expression of indicated genes in CD4+T cells relative to untreated cells following silencing with control (NC) or RNASET2 siRNA. Results are from 4 healthy donors. The right column depicts differential gene expression in IFN- ⁇ secreting compared to non-secreting CD4+T cells.
  • FIG. 43 depicts in accordance with various embodiments of the invention, clinical disease parameters associated with level of ICAM1 expression (measured by RNA-seq) for 71 CD patients based upon IgG ASCA sero-positivity (left panel) or pre-op therapeutic failure of anti-TNF (middle panel) or thiopurine (right panel).
  • RNASET2 RNASET2
  • sequences related to RNASET2 are also incorporated by reference in their entirety as though fully set forth via the rs number disclosed.
  • technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 3 rd ed., Revised , J. Wiley & Sons (New York, N.Y. 2006); and Sambrook et al., Molecular Cloning: A Laboratory Manual 4 th ed ., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y.
  • Bio sample means any biological material from which nucleic acids and/or proteins can be obtained.
  • the term encompasses whole blood, peripheral blood, plasma, serum, saliva, mucus, urine, semen, lymph, fecal extract, cheek swab, cells or other bodily fluid or tissue, including but not limited to tissue obtained through surgical biopsy or surgical resection.
  • a sample can be obtained through primary patient derived cell lines, or archived patient samples in the form of preserved samples, or fresh frozen samples.
  • Treatment and “treating” as used herein refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition, prevent the pathologic condition, pursue or obtain good overall survival, or lower the chances of the individual developing the condition even if the treatment is ultimately unsuccessful.
  • Those in need of treatment include those already with the condition as well as those prone to have the condition or those in whom the condition is to be prevented.
  • SNP single nucleotide polymorphism
  • “Risk variant” as used herein refers to an allele, whose presence is associated with an increase in susceptibility to an inflammatory bowel disease, including but not limited to Crohn's Disease, Ulcerative Colitis and Medically Refractory-Ulcerative Colitis, relative to an individual who does not have the risk variant.
  • IBD Inflammatory Bowel Disease
  • CD Crohn's Disease
  • UC Ulcerative Colitis
  • MR-UC Magnetic Refractive Ulcerative Colitis
  • IBD includes “CD”, “UC” and/or “MR-UC”.
  • ANCA anti-neutrophil cytoplasmic antibodies
  • OptC means outer membrane protein C.
  • eQTL means expression quantitative trait loci.
  • mQTL means methylation quantitative trait loci.
  • Non-limiting examples of RNASET2 SNPs are rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 and rs9366093.
  • RNASET2 RNASET2
  • TL1A TL1A
  • IFN- ⁇ a biomarker of disease severity in a patient population
  • selecting the patient population for anti-TL1A therapy Further described are methods of treating these patient populations.
  • RNASET2 (ribonuclease T2) encodes an extracellular RNase and is the only member of the human Rh/T2/S family of acid ribonucleases (acid hydrolyses), which are only active in acidic pH.
  • the optimal activity of RNASET2 is at pH 5 and it has a preferential cleavage of poly-A and poly-U. It contains two regions with catalytic function and demonstrates a cleavage preference near adenylic acid followed by guanylic acid.
  • Three isoforms have been detected for RNASET2, the 27KD, 31KD and 36KD isoforms. The 27KD and 31KD isoforms are thought to result from proteolytic cleavage of the 36KD isoform.
  • RNASET2 is highly conserved among the phyla, from viruses to humans, suggesting an important evolutionary function.
  • TL1A is a tumor necrosis factor family member expressed primarily on activated cells of the immune system, such as monocytes, macrophages, and dendritic cells, following stimulation by immune complexes or through interaction with enteric microorganisms. TL1A expression is enhanced in inflammatory bowel disease and higher TL1A levels are associated with disease severity.
  • Genome-Wide Association Studies GWAS have identified TNFSF15 SNPs to be associated with IBD. Studies have shown that neutralizing TL1A antibodies attenuate colitis in murine colitis models, while constitutive TL1A expression depicts a worsened murine ileo-cecal inflammation and intestinal fibrostenosis.
  • IFN- ⁇ plays a key role in the generation and perpetuation of mucosal inflammation in IBD.
  • TL1A augments IL-12/IL-18-mediated IFN- ⁇ secretion in PB T cells.
  • RNASET2 an IBD susceptibility gene, as a component of TL1A-mediated enhancement of IFN- ⁇ production.
  • functional variants of RNASET2 are associated with a more ‘severe’ CD phenotype characterized by one or more disease flares and stricturing/penetrating disease behavior.
  • RNASET2 serves as a therapeutic biomarker associated with severe disease pathobiology and allows for the identification of a patient population most likely to benefit from therapy targeted to the TL1A-driven inflammatory pathway.
  • the TNFSF15 disease-associated variants are correlated with increased and sustained expression of TL1A.
  • TNFSF15 has been identified and confirmed in GWAS as an IBD-associated gene and is believed to play a role in modulating the location and severity of intestinal inflammation, as well as development of stricturing disease.
  • Transgenic mice with constitutive expression of TL1A developed intestinal inflammation along with ileal and colonic fibrosis, which was reversed by anti-TL1A treatment.
  • UC In UC, there is a strong association between development of a medically refractory disease and the TL1A locus.
  • TL1A is an important pro-inflammatory cytokine associated with IBD pathogenesis, the molecular pathways underlying enhanced cytokine secretion and inflammation were poorly understood.
  • RNASET2 a component of TL1A-mediated enhancement of IFN- ⁇ production.
  • RNASET2 disease risk SNPs demonstrate a functional association of RNASET2 disease risk SNPs with decreased expression and hyper-methylation in T cells isolated from CD patients, as well as, an association with clinical parameters suggestive of complicated/resistant disease behavior and rapid recurrence of disease.
  • the inventors show the regulatory potential for ETS TF in modulating RNASET2 expression and the involvement of homotypic T cell aggregation via ICAM1 as a component of RNASET2 mediated up-regulation of IFN- ⁇ production.
  • the data distinguish RNASET2 as a potential therapeutic biomarker and identify unique pathways for additional therapeutic modulation within a defined IBD population.
  • RNASET2 RNA-binding protein
  • RNASET2 IBD risk alleles identified through GWAS in samples isolated from the peripheral T cells and small bowel surgical resections.
  • Significant RNASET2 eQTL (rs429083) has been described in a recent report that measured autoimmune associated risk variants in whole thymic tissue samples. This SNP demonstrated the most significant eQTL in the data, as well.
  • the study provides clinically relevant evidence that decreased expression levels of RNASET2 were correlated in CD patients with clinical parameters suggestive of complicated and resistant disease.
  • CD patients carrying the RNASET2 disease risk SNPs displayed an increase in development of stricturing/penetrating disease behavior.
  • RNASET2 expression was significantly lower in T cells isolated from CD patients exhibiting one or more disease flares per year. Similarly, RNASET2 expression is decreased in small bowel mucosal samples, as well as, in peripheral samples from medically refractory CD patients (9 out of 11 which were non-responsive to anti-TNF therapy), requiring surgical intervention for disease management. Consistent with the finding, a recent study reported significant RNASET2 eQTL in whole blood from patients resistant to anti-TNF therapy. Moreover, RNASET2 disease-associated SNPs correlated with therapeutic failure of anti-TNF therapy, and intestinal resection of >40 cm clinical characteristic of overall disease severity.
  • RNASET2 disease risk SNP was associated with a faster time to reoperation.
  • RNASET2 disease associated SNPs were associated in patients with endoscopic recurrence characterized by a more severe (>2) Rutgeerts score, which without being bound to any particular theory, can be predictive for early clinical recurrence and need for reoperation.
  • RNASET2 expression The transcriptional regulatory regions and binding factors modulating RNASET2 expression are likewise poorly defined.
  • the majority of disease associated variants identified by GWAS reside within regulatory non-coding regions corresponding to promoters or enhancer sequences. Without being bound to any particular theory, studies suggest that alteration in transcriptional regulation via disruption of transcription factor binding sites may play a role in the disease process.
  • the inventors utilized TF motif analysis to prioritize and identify from the large number of variants demonstrating eQTL and mQTL a prospective regulatory SNP.
  • the rs2149092 disease associated SNP alters the conserved ETS consensus binding sequence and likely disrupts binding of multiple overlapping TF binding sites including IRF4, SPI1 and ELF1.
  • RNASET2 the cell adhesion molecule
  • ICAM1A the cell adhesion molecule
  • Homotypic T-T aggregates have been shown to promote synaptic-based cytokine delivery of IFN- ⁇ and IL2 from one T cell to another, resulting in IL-2 receptor ligation and subsequent STATS phosphorylation.
  • the inventors demonstrate that enhanced cellular aggregation is a hallmark of IFN- ⁇ producing cells and TL1A-stimulation increases the number and size of the cellular aggregates.
  • RNASET2 may act through the integrin signaling pathway to modulate downstream IFN- ⁇ secretion.
  • RNASET2 a novel functional and biological relationship between two IBD susceptibility genes, TNFSF15 and RNASET2.
  • the inventors provide evidence that decreased RNASET2 expression is functionally implicated in both the TL1A driven pro-inflammatory cytokine production by activated T cells and functionally associated with the RNASET2 IBD susceptibility variants.
  • the present study demonstrates an association between decreased RNASET2 expression and a more severe form of IBD inflammation, which without wishing to be bound by any particular theory, we believe underlie disease pathology triggered by TL1A mediated pathways. Decreased expression and altered epigenetic DNA methylation of RNASET2 characterize a subset of IBD patients with a more severe disease phenotype.
  • RNASET2 disease risk SNPs demonstrate a functional association of RNASET2 disease risk SNPs with decreased expression and hyper-methylation in T cells isolated from CD patients, as well as, an association with clinical parameters suggestive of complicated/resistant disease behavior and rapid recurrence of disease.
  • the inventors show the regulatory potential for ETS TF in modulating RNASET2 expression and the involvement of homotypic T cell aggregation via ICAM1 as a component of RNASET2 mediated upregulation of IFN- ⁇ production.
  • the data distinguish RNASET2 as a potential therapeutic biomarker and identify unique pathways for additional therapeutic modulation within a defined IBD population.
  • RNASET2 expression serves as a novel disease biomarker of a more severe form of inflammation identifying a patient population not responsive to current treatment strategies, whom may benefit from alternate RNASET2 mediated therapeutic approaches.
  • RNASET2 associated SNPs in an IBD patient cohort.
  • the inventors have identified RNASET2 associated SNPs in a CD patient cohort.
  • the inventors have identified RNASET2 associated SNPs in a UC patient cohort.
  • the inventors have identified RNASET2 associated SNPs in a MR-UC patient cohort.
  • the SNPs were associated with disease location, disease behavior and need for surgery.
  • the inventors have further identified RNASET2 as a biomarker for disease severity and associated RNASET2 risk SNPs in an IBD patient cohort.
  • RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a CD patient cohort.
  • RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a UC patient cohort.
  • RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a MR-UC patient cohort.
  • the inventors demonstrate a correlation between RNASET2, TL1A expression and IFN- ⁇ secretion.
  • the present invention is based, at least in part, on these findings.
  • the present invention addresses the need in the art for methods of diagnosing patients with IBD and identifying patients in need of treatment, using RNASET2, TL1A and/or IFN- ⁇ .
  • the invention further provides a process for patient identification and/or stratification.
  • inflammatory bowel disease in a subject, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; and diagnosing IBD in the subject based on the presence of one or more risk variants at the RNASET2 gene.
  • inflammatory bowel disease is Crohn's disease, ulcerative colitis or medically refractive ulcerative colitis.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093.
  • the subject is diagnosed with IBD if 2, 3, 4, 5 or 6 RNASET2 risk variants as described herein are present.
  • the risk allele for rs2149085 is the T allele.
  • the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085.
  • the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085, as described herein are present.
  • the presence of a greater number of risk variants in the sample indicates that the subject is in greater need of treatment.
  • the detection of RNASET2 risk variants is indicative of the need for treatment in the subject.
  • the subject is identified as needing anti-TL1A therapy.
  • the subject diagnosed with IBD demonstrates therapeutic failure of thiopurine and anti-TNF therapy.
  • the subject diagnosed with IBD is determined to need surgical intervention.
  • the surgical intervention is intestinal resection.
  • RNASET2 risk variants SNP SEQ ID NO: rs1819333 1 rs2149092 2 rs9355610 3 rs2149085 4 rs1410295 5 rs9366093 6
  • the method of diagnosing inflammatory bowel disease (IBD) in a subject described herein comprises determining the expression level of RNASET2, TL1A and/or IFN- ⁇ .
  • a subject with decreased RNASET2, and/or increased TL1A and/or IFN- ⁇ levels is diagnosed with IBD.
  • inflammatory bowel disease is Crohn's disease.
  • inflammatory bowel disease is ulcerative colitis.
  • inflammatory bowel disease is medically refractive ulcerative colitis.
  • inflammatory bowel disease is a CD patient who required surgical intervention for disease management.
  • a subject with decreased RNASET2, and/or increased TL1A and/or IFN- ⁇ levels is identified as a subject in need of a treatment that increases RNASET2, and/or decreases TL1A and/or IFN- ⁇ .
  • the subject is identified as needing anti-TL1A therapy.
  • the subject is identified as needing a treatment that causes an increase in RNASET2.
  • the subject is identified as needing a treatment that causes a decrease in IFN- ⁇ and/or TL1A.
  • RNASET2 risk variants and/or RNASET2, TL1A and/or IFN- ⁇ expression levels can be accomplished by analyzing nucleic acids of a biological sample from the subject.
  • a variety of apparatuses and/or methods including, without limitation, polymerase chain reaction based analysis, sequence analysis and electrophoretic analysis can be used to detect RNASET2 risk variants.
  • the expression levels of RNASET2, TL1A and/or IFN- ⁇ can be detected using a variety of apparatuses and/or methods, including, without limitation, quantitative PCR, northern blot and microarrays.
  • nucleic acid means a polynucleotide such as a single or double-stranded DNA or RNA molecule including, for example, genomic DNA, cDNA and mRNA.
  • nucleic acid encompasses nucleic acid molecules of both natural and synthetic origin as well as molecules of linear, circular or branched configuration representing either the sense or antisense strand, or both, of a native nucleic acid molecule.
  • determining the expression level of RNASET2, TL1A and/or IFN- ⁇ can be accomplished by analyzing the proteins of a biological sample from the subject.
  • a variety of apparatuses and/or methods, including, without limitation, ELISA, immunohistochemistry, and western blot can be used to detect RNASET2, TL1A and/or IFN- ⁇ expression levels.
  • Various embodiments of the present invention also provide for a method of diagnosing medically refractive ulcerative colitis (MR-UC), comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2; and diagnosing MR-UC in the subject based on the presence of one or more risk variants at the RNASET2 gene.
  • the one or more risk variants at the RNASET2 gene are rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093.
  • the risk allele for rs2149085 is the T allele.
  • the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085.
  • the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present.
  • the subject diagnosed with MR-UC demonstrates therapeutic failure of thiopurine and anti-TNF therapy.
  • the subject diagnosed with MR-UC is determined to need surgical intervention.
  • the surgical intervention is intestinal resection.
  • the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • the methods further comprise determining the level of methylation of RNASET2, and diagnosing IBD in a subject who has an increase in RNASET2 methylation.
  • the level of RNASET2 methylation is determined to diagnose a subject with MR-UC.
  • the subject is identified as needing a treatment that causes a decrease in RNASET2 methylation.
  • the subject is identified as needing an anti-TL1A therapy.
  • MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine.
  • the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • the subject is identified as needing a treatment that mimics, RNASET2, TL1A and/or IFN- ⁇ . In other embodiments, the subject is identified as needing a treatment that modulates RNASET2, TL1A and/or IFN- ⁇ . In some other embodiments, the subject is identified as needing a treatment that targets RNASET2, TL1A and/or IFN- ⁇ . In yet other embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the disease is IBD.
  • the disease is CD.
  • the disease is UC.
  • the disease is MR-UC.
  • the subject diagnosed is a CD patients who required surgical intervention for disease management.
  • the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2. In other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2 in peripheral and mucosal tissues. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with DNA hypermethylation in patients requiring surgical intervention for disease management. In yet other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with therapeutic failure of thiopurine and/or anti-TNF therapy. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with ANCA sero-positivity. In various other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with increased overall length of intestinal resection.
  • RNASET2 a subject with inflammatory bowel disease for treatment
  • TL1A and/or IFN- ⁇ a subject with inflammatory bowel disease for treatment
  • identifying the subject in need of treatment as a subject with decreased RNASET2, and/or increased TL1A and/or IFN- ⁇ levels a subject with decreased RNASET2, and/or increased TL1A and/or IFN- ⁇ levels.
  • the inflammatory bowel disease is Crohn's disease.
  • the inflammatory bowel disease is ulcerative colitis.
  • the inflammatory bowel disease is medically refractive ulcerative colitis.
  • the subject is identified as needing a treatment that causes an increase in RNASET2.
  • the subject is identified as needing a treatment that causes a decrease in TL1A and/or IFN- ⁇ . In certain embodiments, the subject is identified as needing anti-TL1A therapy. In some embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ . In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • Patient Risk Stratification as used herein means the process of separating subjects into risk groups in need of treatment.
  • RNASET2 is indicative of a patient having IBD, in need of treatment.
  • a decrease in RNASET2 is indicative of a patient having CD, in need of treatment.
  • the patient is a CD patient who requires surgical intervention for disease management.
  • a decrease in RNASET2 is indicative of a patient having UC, in need of treatment.
  • a decrease in RNASET2 is indicative of a patient having MR-UC in need of treatment.
  • an increase in TL1A and/or IFN- ⁇ is indicative of a patient having IBD, in need of treatment.
  • an increase in TL1A and/or IFN- ⁇ is indicative of a patient having CD, in need of treatment.
  • an increase in TL1A and/or IFN- ⁇ is indicative of a patient having UC, in need of treatment.
  • an increase in TL1A and/or IFN- ⁇ is indicative of a patient having MR-UC, in need of treatment.
  • a decrease in RNASET2, an increase in TL1A, and an increase in IFN- ⁇ is indicative of a subject having IBD, in need of treatment.
  • a decrease in RNASET2, an increase in TL1A, and an increase in IFN- ⁇ is indicative of a subject having CD, in need of treatment.
  • a decrease in RNASET2, an increase in TL1A, and an increase in IFN- ⁇ is indicative of a subject having UC, in need of treatment.
  • a decrease in RNASET2, an increase in TL1A, and an increase in IFN- ⁇ is indicative of a subject having MR-UC, in need of treatment.
  • detection of the genes provides a guide for the treatment of the subject.
  • the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the process of patient risk stratification to identify a subject in need of treatment is relative to a healthy individual who has been previously treated.
  • the process of patient risk stratification to identify a subject in need of treatment is relative to a medically responsive individual.
  • MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine.
  • the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • the process for subject identification and/or stratification described herein comprises determining the presence of one or more risk variants.
  • the one or more risk variants comprise rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093.
  • the process comprises identifying the subject with IBD in need of treatment if 2, 3, 4, 5 or 6 RNASET2 risk variants as described herein are present.
  • the risk allele for rs2149085 is the T allele.
  • the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085.
  • the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present.
  • the detection of the risk variants in the biological sample stratifies the subject into a group needing treatment. In other embodiments, the presence of a greater number of risk variants in the sample indicates that the subject is in greater need of treatment.
  • the detection of RNASET2 risk variants is indicative of the need for treatment in the subject.
  • the subject is identified as needing anti-TL1A therapy.
  • the subject identified as needing RNASET2 mediated therapy such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • RNASET2 risk variants can be accomplished by analyzing nucleic acids of a biological sample from the subject, as discussed herein.
  • the process for subject identification and/or stratification described herein further comprises assaying the sample to detect the level of RNASET2 methylation, relative to a healthy individual.
  • a subject with an increased level of RNASET2 methylation is identified as a subject in need of treatment.
  • the sample is assessed for the level of RNASET2 methylation and one or more RNASET2 risk variants.
  • a subject who has an increase in RNASET2 methylation and the presence of one or more RNASET2 risk variants is identified as a subject in need of treatment.
  • the sample is assessed for the level of RNASET2 methylation and the expression levels of RNASET2, TL1A and/or IFN- ⁇ .
  • a subject who has an increase in RNASET2 methylation and a decrease in RNASET2, an increase in TL1A and/or IFN- ⁇ is identified as a subject in need of treatment.
  • the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the subject is identified as needing anti-TL1A therapy. In some embodiments, the subject is identified as needing a treatment that causes a decrease in RNASET2 methylation. In various other embodiments, the detection of an increase in RNASET2 methylation is indicative of a patient with severe CD requiring surgery. In a further embodiment, the subject is identified as needing a treatment that comprises colectomy and/or anti-TL1A therapy.
  • the process for subject identification and/or stratification described herein can further comprise assaying the sample to detect an increase or decrease of at least one microbial antigen (serological factor), relative to a healthy individual.
  • the microbial antigens (serological factors) assessed comprise ANCA, ASCA, OmpC, I2 and CBir.
  • the sample is assessed for one or more microbial antigens (serological factors) and one or more RNASET2 risk variants.
  • a subject who has one or more risk serological factors and the presence of one or more RNASET2 risk variants is identified as a subject in need of treatment.
  • the sample is assessed for one or more risk serological factors and the expression levels of RNASET2, TL1A and/or IFN- ⁇ .
  • a subject who has one or more risk serological factors and a decrease in RNASET2, an increase in TL1A and/or IFN- ⁇ is identified as a subject in need of treatment.
  • the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the treatment is anti-TL1A therapy.
  • the treatment is an RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • the subject identified with IBD demonstrates therapeutic failure of thiopurine and anti-TNF therapy.
  • the subject identified with IBD is determined to need surgical intervention.
  • the surgical intervention is intestinal resection.
  • Various embodiments of the present invention also provide for a method of selecting surgery for a subject having Inflammatory Bowel Disease, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; diagnosing MR-UC in the subject based on the presence of one or more risk variants at the RNASET2 gene; and selecting surgery for the subject diagnosed with MR-UC.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093.
  • the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085.
  • the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present.
  • the method further comprises determining the level of methylation of RNASET2.
  • a subject with an increased level of RNASET2 methylation is identified as a subject in need of surgery.
  • the subject with an increase in RNASET2 methylation and the presence of one or more risk variants at the RNASET2 gene is identified as a subject in need of surgery.
  • the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2. In other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2 in peripheral and mucosal tissues. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with DNA hypermethylation in patients requiring surgical intervention for disease management. In yet other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with therapeutic failure of thiopurine and/or anti-TNF therapy. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with ANCA sero-positivity. In various other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with increased overall length of intestinal resection.
  • Various embodiments of the present invention provide for a method of selecting a therapy for a subject having Inflammatory Bowel Disease, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; diagnosing medically refractive ulcerative colitis (MR-UC) in the subject based on the presence of one or more risk variants at the RNASET2 gene; and selecting surgery as the therapy and not selecting thiopurine or anti-TNF as the therapy for the subject diagnosed with MR-UC.
  • MR-UC medically refractive ulcerative colitis
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093.
  • the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof.
  • the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085.
  • the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present.
  • the method further comprises determining the level of methylation of RNASET2, wherein increased methylation is indicative of a subject requiring surgical intervention.
  • MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine.
  • the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • RNASET2 inflammatory bowel disease
  • identifying the subject with IBD as a subject with increased RNASET2 methylation In other embodiments, the method comprises identifying the subject with IBD as a subject who has increased RNASET2 methylation and who has the presence of one or more risk variants at the RNASET2 gene.
  • the method comprises determining the expression level of RNASET2, TL1A and/or IFN- ⁇ ; and diagnosing the subject with IBD if the subject has a decrease in RNASET2, an increase in TL1A, an increase in IFN- ⁇ and/or an increase in RNASET2 methylation.
  • the method comprises determining the level of methylation of RNASET2; and identifying the subject in need of treatment as a subject with increased RNASET2 methylation.
  • the expression level of RNASET2, TL1A and/or IFN- ⁇ and the level of methylation of RNASET2 are determined, to identify a subject with inflammatory bowel disease in need of treatment.
  • the method comprises determining a subject in need of treatment as a subject who has a decrease in RNASET2, an increase in TL1A, an increase in IFN- ⁇ , and/or an increase in RNASET2 methylation.
  • the method comprises determining the presence or absence of one or more risk variants at the RNASET2 gene and identifying a subject with IBD in need of treatment as a subject who has an increase in RNASET2 methylation and the presence of one or more RNASET2 risk variants.
  • an increase in methylation is indicative of a subject requiring surgical intervention.
  • an increase in RNASET2 methylation is indicative of requiring surgical intervention.
  • Various methods to detect levels of methylation include, but are not limited to the following assays, mass spectrometry, methylation-specific PCR (MSP), whole genome bisulfite sequencing, (BS-Seq), the HELP assay, ChIP-on-chip assays, restriction landmark genomic scanning, methylated DNA immunoprecipitation (MeDIP, MeDIP-chip, MeDIPseq), pyrosequencing of bisulfite treated DNA, molecular break light assay for DNA adenine methyltransferase activity, methyl sensitive southern blotting, separate native DNA into methylated and unmethylated fractions using MethylCpG Binding Proteins (MBPs) and/or Methyl Binding Domain (MBD), MethylationEPIC BeadChip, Illumina Infinium Methylation 450 BeadChip, High Resolution Melt Analysis (HRM or HRMA), and/or ancient DNA methylation reconstruction.
  • MSP methylation-
  • inflammatory bowel disease provides for the treatment of a subject diagnosed with inflammatory bowel disease (IBD) by the method comprising obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; and diagnosing IBD in the subject based on the presence of one or more risk variants at the RNASET2 gene.
  • IBD inflammatory bowel disease
  • inflammatory bowel disease is Crohn's disease, ulcerative colitis or medically refractive ulcerative colitis.
  • MR-UC medically refractive ulcerative colitis
  • MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy and thiopurine therapy.
  • the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ .
  • treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN- ⁇ can comprise antibodies and or silencing oligonucleotides.
  • the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1.
  • RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • the steps involved in the current invention comprise obtaining a biological sample from a subject.
  • the biological sample may be obtained either through surgical biopsy or surgical resection.
  • a sample can be obtained through primary patient derived cell lines, or archived patient samples in the form of FFPE (Formalin fixed, paraffin embedded) samples, or fresh frozen samples.
  • a sample may also comprise whole blood, peripheral blood, plasma, serum, saliva, cheek swab, or other bodily fluid or tissue.
  • the sample comprises tissue from the large and/or small intestine.
  • the large intestine sample comprises the cecum, colon (the ascending colon, the transverse colon, the descending colon, and the sigmoid colon), rectum and/or the anal canal.
  • the small intestine sample comprises the duodenum, jejunum, and/or the ileum.
  • Nucleic acid or protein samples derived from the biological sample (i.e., tissue and/or cells) of a subject that can be used in the methods of the invention can be prepared by means well known in the art. For example, surgical procedures or needle biopsy aspiration can be used to collect the biological samples from a subject. In some embodiments, it is important to enrich and/or purify the abnormal tissue and/or cell samples from the normal tissue and/or cell samples. In other embodiments, the abnormal tissue and/or cell samples can then be microdissected to reduce the amount of normal tissue contamination prior to extraction of genomic nucleic acid or pre-RNA for use in the methods of the invention. Such enrichment and/or purification can be accomplished according to methods well-known in the art, such as needle microdissection, laser microdissection, fluorescence activated cell sorting, and immunological cell sorting.
  • RNASET2 Analysis of the nucleic acid and/or protein from an individual may be performed using any of various techniques.
  • assaying gene expression levels for RNASET2 comprises northern blot, reverse transcription PCR, real-time PCR, serial analysis of gene expression (SAGE), DNA microarray, tiling array, RNA-Seq, or a combination thereof.
  • SAGE serial analysis of gene expression
  • DNA microarray DNA microarray
  • tiling array RNA-Seq
  • the gene expression levels for RNASET2, TL1A and/or IFN- ⁇ are assayed.
  • the level of RNASET2 methylation is determined.
  • methods and systems to detect protein expression include but are not limited to ELISA, immunohistochemistry, western blot, flow cytometry, fluorescence in situ hybridization (FISH), radioimmuno assays, and affinity purification.
  • the analysis of gene expression levels may involve amplification of an individual's nucleic acid by the polymerase chain reaction.
  • Use of the polymerase chain reaction for the amplification of nucleic acids is well known in the art (see, for example, Mullis et al. (Eds.), The Polymerase Chain Reaction, Birkhauser, Boston, (1994)).
  • Quantitative amplification involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction.
  • Detailed protocols for quantitative PCR are provided in Innis, et al. (1990) PCR Protocols, A Guide to Methods and Applications , Academic Press, Inc. N.Y.). Measurement of DNA copy number at microsatellite loci using quantitative PCR analysis is described in Ginzonger, et al. (2000) Cancer Research 60:5405-5409.
  • the known nucleic acid sequence for the genes is sufficient to enable one of skill in the art to routinely select primers to amplify any portion of the gene.
  • Fluorogenic quantitative PCR may also be used in the methods of the invention. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green.
  • ligase chain reaction (LCR) (see Wu and Wallace (1989) Genomics 4: 560, Landegren, et al. (1988) Science 241:1077, and Barringer et al. (1990) Gene 89: 117), transcription amplification (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), self-sustained sequence replication (Guatelli, et al. (1990) Proc. Nat. Acad. Sci. USA 87: 1874), dot PCR, and linker adapter PCR, etc.
  • LCR ligase chain reaction
  • a DNA sample suitable for hybridization can be obtained, e.g., by polymerase chain reaction (PCR) amplification of genomic DNA, fragments of genomic DNA, fragments of genomic DNA ligated to adaptor sequences or cloned sequences.
  • Computer programs that are well known in the art can be used in the design of primers with the desired specificity and optimal amplification properties, such as Oligo version 5.0 (National Biosciences).
  • PCR methods are well known in the art, and are described, for example, in Innis et al., eds., 1990, PCR Protocols: A Guide to Methods And Applications, Academic Press Inc., San Diego, Calif. It will be apparent to one skilled in the art that controlled robotic systems are useful for isolating and amplifying nucleic acids and can be used.
  • the nucleic acid samples derived from a subject used in the methods of the invention can be hybridized to arrays comprising probes (e.g., oligonucleotide probes) in order to identify RNASET2, TL1A and/or IFN- ⁇ and in instances wherein a housekeeping gene expression is also to be assessed, comprising probes in order to identify selected housekeeping genes.
  • the probes used in the methods of the invention comprise an array of probes that can be tiled on a DNA chip (e.g., SNP oligonucleotide probes).
  • Hybridization and wash conditions used in the methods of the invention are chosen so that the nucleic acid samples to be analyzed by the invention specifically bind or specifically hybridize to the complementary oligonucleotide sequences of the array, preferably to a specific array site, wherein its complementary DNA is located.
  • the complementary DNA can be completely matched or mismatched to some degree as used, for example, in Affymetrix oligonucleotide arrays.
  • the single-stranded synthetic oligodeoxyribonucleic acid DNA probes of an array may need to be denatured prior to contact with the nucleic acid samples from a subject, e.g., to remove hairpins or dimers which form due to self-complementary sequences.
  • Optimal hybridization conditions will depend on the length of the probes and type of nucleic acid samples from a subject.
  • General parameters for specific (i.e., stringent) hybridization conditions for nucleic acids are described in Sambrook and Russel, Molecular Cloning: A Laboratory Manual 4 th ed ., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y. 2012); Ausubel et al., eds., 1989, Current Protocols in Molecules Biology, Vol. 1, Green Publishing Associates, Inc., John Wiley & Sons, Inc., New York, at pp. 2.10.1-2.10.16.
  • Exemplary useful hybridization conditions are provided in, e.g., Tijessen, 1993, Hybridization with Nucleic Acid Probes, Elsevier Science Publishers B. V. and Kricka, 1992, Nonisotopic DNA Probe Techniques, Academic Press, San Diego, Calif.
  • DNA arrays can be used to determine the expression levels of genes, by measuring the level of hybridization of the nucleic acid sequence to oligonucleotide probes that comprise complementary sequences.
  • oligonucleotide probes i.e., nucleic acid molecules having defined sequences
  • a set of nucleic acid probes, each of which has a defined sequence is immobilized on a solid support in such a manner that each different probe is immobilized to a predetermined region.
  • the set of probes forms an array of positionally-addressable binding (e.g., hybridization) sites on a support.
  • Each of such binding sites comprises a plurality of oligonucleotide molecules of a probe bound to the predetermined region on the support.
  • each probe of the array is preferably located at a known, predetermined position on the solid support such that the identity (i.e., the sequence) of each probe can be determined from its position on the array (i.e., on the support or surface).
  • Microarrays can be made in a number of ways, of which several are described herein. However produced, microarrays share certain characteristics, they are reproducible, allowing multiple copies of a given array to be produced and easily compared with each other.
  • the microarrays are made from materials that are stable under binding (e.g., nucleic acid hybridization) conditions.
  • the microarrays are preferably small, e.g., between about 1 cm 2 and 25 cm 2 , preferably about 1 to 3 cm 2 .
  • both larger and smaller arrays are also contemplated and may be preferable, e.g., for simultaneously evaluating a very large number of different probes.
  • Oligonucleotide probes can be synthesized directly on a support to form the array.
  • the probes can be attached to a solid support or surface, which may be made, e.g., from glass, plastic (e.g., polypropylene, nylon), polyacrylamide, nitrocellulose, gel, or other porous or nonporous material.
  • the set of immobilized probes or the array of immobilized probes is contacted with a sample containing labeled nucleic acid species so that nucleic acids having sequences complementary to an immobilized probe hybridize or bind to the probe. After separation of, e.g., by washing off, any unbound material, the bound, labeled sequences are detected and measured. The measurement is typically conducted with computer assistance.
  • DNA array technologies have made it possible to determine the expression level of RNASET2, TL1A and/or IFN- ⁇ , housekeeping genes and the methylation state of RNASET2.
  • high-density oligonucleotide arrays are used in the methods of the invention. These arrays containing thousands of oligonucleotides complementary to defined sequences, at defined locations on a surface can be synthesized in situ on the surface by, for example, photolithographic techniques (see, e.g., Fodor et al., 1991, Science 251:767-773; Pease et al., 1994, Proc. Natl. Acad. Sci. U.S.A. 91:5022-5026; Lockhart et al., 1996, Nature Biotechnology 14:1675; U.S. Pat. Nos.
  • Another method for attaching the nucleic acids to a surface is by printing on glass plates, as is described generally by Schena et al. (1995, Science 270:467-470).
  • Other methods for making microarrays e.g., by masking (Maskos and Southern, 1992, Nucl. Acids. Res. 20:1679-1684), may also be used.
  • oligonucleotides e.g., 15 to 60-mers
  • the array produced can be redundant, with several oligonucleotide molecules corresponding to each informative locus of interest (e.g., SNPs, RFLPs, STRs, etc.).
  • One exemplary means for generating the oligonucleotide probes of the DNA array is by synthesis of synthetic polynucleotides or oligonucleotides, e.g., using N-phosphonate or phosphoramidite chemistries (Froehler et al., 1986, Nucleic Acid Res. 14:5399-5407; McBride et al., 1983, Tetrahedron Lett. 24:246-248). Synthetic sequences are typically between about 15 and about 600 bases in length, more typically between about 20 and about 100 bases, most preferably between about 40 and about 70 bases in length.
  • synthetic nucleic acids include non-natural bases, such as, but by no means limited to, inosine.
  • nucleic acid analogues may be used as binding sites for hybridization.
  • An example of a suitable nucleic acid analogue is peptide nucleic acid (see, e.g., Egholm et al., 1993, Nature 363:566-568; U.S. Pat. No. 5,539,083).
  • the hybridization sites i.e., the probes
  • the probes are made from plasmid or phage clones of regions of genomic DNA corresponding to SNPs or the complement thereof.
  • the size of the oligonucleotide probes used in the methods of the invention can be at least 10, 20, 25, 30, 35, 40, 45, or 50 nucleotides in length.
  • hybridization stringency condition e.g., the hybridization temperature and the salt concentrations, may be altered by methods that are well known in the art.
  • the high-density oligonucleotide arrays used in the methods of the invention comprise oligonucleotides corresponding to RNASET2, TL1A and/or IFN- ⁇ and housekeeping genes.
  • the oligonucleotides correspond to methylated RNASET2.
  • the oligonucleotide probes may comprise DNA or DNA “mimics” (e.g., derivatives and analogues) corresponding to a portion of each informative locus of interest (e.g., SNPs, RFLPs, STRs, etc.) in a subject's genome.
  • oligonucleotide probes can be modified at the base moiety, at the sugar moiety, or at the phosphate backbone.
  • Exemplary DNA mimics include, e.g., phosphorothioates.
  • a plurality of different oligonucleotides may be used that are complementary to the sequences of sample nucleic acids.
  • a single informative locus of interest e.g., SNPs, RFLPs, STRs, etc.
  • a single informative locus of interest e.g., SNPs, RFLPs, STRs, etc.
  • about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more different oligonucleotides can be used.
  • Each of the oligonucleotides for a particular informative locus of interest may have a slight variation in perfect matches, mismatches, and flanking sequence around the SNP.
  • the probes are generated such that the probes for a particular informative locus of interest comprise overlapping and/or successive overlapping sequences which span or are tiled across a genomic region containing the target site, where all the probes contain the target site.
  • overlapping probe sequences can be tiled at steps of a predetermined base interval, e. g. at steps of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 bases intervals.
  • the assays can be performed using arrays suitable for use with molecular inversion probe protocols such as described by Wang et al. (2007) Genome Biol. 8, R246.
  • cross-hybridization among similar probes can significantly contaminate and confuse the results of hybridization measurements.
  • Cross-hybridization is a particularly significant concern in the detection of SNPs since the sequence to be detected (i.e., the particular SNP) must be distinguished from other sequences that differ by only a single nucleotide.
  • Cross-hybridization can be minimized by regulating either the hybridization stringency condition and/or during post-hybridization washings.
  • the probes used in the methods of the invention are immobilized (i.e., tiled) on a glass slide called a chip.
  • a DNA microarray can comprises a chip on which oligonucleotides (purified single-stranded DNA sequences in solution) have been robotically printed in an (approximately) rectangular array with each spot on the array corresponds to a single DNA sample which encodes an oligonucleotide.
  • the process comprises, flooding the DNA microarray chip with a labeled sample under conditions suitable for hybridization to occur between the slide sequences and the labeled sample, then the array is washed and dried, and the array is scanned with a laser microscope to detect hybridization.
  • RNASET2, TL1A and/or IFN- ⁇ or housekeeping genes for which probes appear on the array (with match/mismatch probes for a single locus of interest or probes tiled across a single locus of interest counting as one locus of interest).
  • RNASET2, TL1A and/or IFN- ⁇ or housekeeping genes being probed per array is determined by the size of the genome and genetic diversity of the subjects species.
  • DNA chips are well known in the art and can be purchased in pre-5 fabricated form with sequences specific to particular species.
  • SNPs and/or DNA copy number can be detected and quantitated using sequencing methods, such as “next-generation sequencing methods” as described further above.
  • the protein, polypeptide, nucleic acid, fragments thereof, or fragments thereof ligated to adaptor regions used in the methods of the invention are detectably labeled.
  • the detectable label can be a fluorescent label, e.g., by incorporation of nucleotide analogues.
  • Other labels suitable for use in the present invention include, but are not limited to, biotin, iminobiotin, antigens, cofactors, dinitrophenol, lipoic acid, olefinic compounds, detectable polypeptides, electron rich molecules, enzymes capable of generating a detectable signal by action upon a substrate, and radioactive isotopes.
  • Radioactive isotopes include that can be used in conjunction with the methods of the invention, but are not limited to, 32P and 14C.
  • Fluorescent molecules suitable for the present invention include, but are not limited to, fluorescein and its derivatives, rhodamine and its derivatives, texas red, 5′carboxy-fluorescein (“FAM”), 2′, 7′-dimethoxy-4′, 5′-dichloro-6-carboxy-fluorescein (“JOE”), N, N, N′, N′-tetramethyl-6-carboxy-rhodamine (“TAMRA”), 6-carboxy-X-rhodamine (“ROX”), HEX, TET, IRD40, and IRD41.
  • Fluorescent molecules which are suitable for use according to the invention further include: cyamine dyes, including but not limited to Cy2, Cy3, Cy3.5, CY5, Cy5.5, Cy7 and FLUORX; BODIPY dyes including but not limited to BODIPY-FL, BODIPY-TR, BODIPY-TMR, BODIPY-630/650, and BODIPY-650/670; and ALEXA dyes, including but not limited to ALEXA-488, ALEXA-532, ALEXA-546, ALEXA-568, and ALEXA-594; as well as other fluorescent dyes which will be known to those who are skilled in the art.
  • Electron rich indicator molecules suitable for the present invention include, but are not limited to, ferritin, hemocyanin and colloidal gold.
  • Two-color fluorescence labeling and detection schemes may also be used (Shena et al., 1995, Science 270:467-470). Use of two or more labels can be useful in detecting variations due to minor differences in experimental conditions (e.g., hybridization conditions). In some embodiments of the invention, at least 5, 10, 20, or 100 dyes of different colors can be used for labeling. Such labeling would also permit analysis of multiple samples simultaneously which is encompassed by the invention.
  • the labeled nucleic acid samples, fragments thereof, or fragments thereof ligated to adaptor regions that can be used in the methods of the invention are contacted to a plurality of oligonucleotide probes under conditions that allow sample nucleic acids having sequences complementary to the probes to hybridize thereto.
  • the hybridization signals can be detected using methods well known to those of skill in the art including, but not limited to, X-Ray film, phosphor imager, or CCD camera.
  • the fluorescence emissions at each site of a transcript array can be, preferably, detected by scanning confocal laser microscopy.
  • a separate scan, using the appropriate excitation line, is carried out for each of the two fluorophores used.
  • a laser can be used that allows simultaneous specimen illumination at wavelengths specific to the two fluorophores and emissions from the two fluorophores can be analyzed simultaneously (see Shalon et al. (1996) Genome Res. 6, 639-645).
  • the arrays are scanned with a laser fluorescence scanner with a computer controlled X-Y stage and a microscope objective. Sequential excitation of the two fluorophores is achieved with a multi-line, mixed gas laser, and the emitted light is split by wavelength and detected with two photomultiplier tubes.
  • RNASET2 RNASET2
  • TL1A RNASET2
  • IFN- ⁇ housekeeping genes
  • the amplification can comprise cloning regions of genomic DNA of the subject.
  • amplification of the DNA regions is achieved through the cloning process.
  • expression vectors can be engineered to express large quantities of particular fragments of genomic DNA of the subject (Sambrook and Russel, Molecular Cloning: A Laboratory Manual 4 th ed ., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y. 2012)).
  • the amplification comprises expressing a nucleic acid encoding a gene, or a gene and flanking genomic regions of nucleic acids, from the subject.
  • RNA pre-messenger RNA
  • RNA pre-messenger RNA
  • the genomic DNA, or pre-RNA, of a subject may be fragmented using restriction endonucleases or other methods. The resulting fragments may be hybridized to SNP probes.
  • a DNA sample of a subject for use in hybridization may be about 400 ng, 500 ng, 600 ng, 700 ng, 800 ng, 900 ng, or 1000 ng of DNA or greater.
  • methods are used that require very small amounts of nucleic acids for analysis, such as less than 400 ng, 300 ng, 200 ng, 100 ng, 90 ng, 85 ng, 80 ng, 75 ng, 70 ng, 65 ng, 60 ng, 55 ng, 50 ng, or less, such as is used for molecular inversion probe (MIP) assays.
  • MIP molecular inversion probe
  • the resulting data can be analyzed using various algorithms, based on well-known methods used by those skilled in the art.
  • the present invention is also directed to a kit to diagnose a subject with IBD
  • the kit is useful for practicing the inventive method of diagnosing a subject and/or identifying a subject in need of treatment.
  • the kit is an assemblage of materials or components, including at least one of the inventive compositions.
  • the kit contains a composition including primers and probes for RNASET2, TL1A and/or IFN- ⁇ , as described above.
  • the kit is configured to detect the gene expression levels of RNASET2 in a sample.
  • the kit is configured to detect the gene expression levels of RNASET2 and/or TL1A in a sample.
  • the kit is configured to detect the gene expression levels of RNASET2, TL1A and/or IFN- ⁇ in a sample.
  • the kit is configured to detect RNASET2 risk variants in a sample.
  • the kit is configured to detect the level of RNASET2 methylation in a sample.
  • the kit is configured particularly for the purpose of assessing mammalian subjects. In another embodiment, the kit is configured particularly for the purpose of assessing human subjects. In further embodiments, the kit is configured for veterinary applications, assessing subjects such as, but not limited to, farm animals, domestic animals, and laboratory animals.
  • Instructions for use may be included in the kit.
  • “Instructions for use” typically include a tangible expression describing the technique to be employed in using the components of the kit to effect a desired outcome, such as to diagnose a subject with IBD and/or identify a subject with IBD in need of treatment.
  • the kit also contains other useful components, such as, primers, diluents, buffers, pipetting or measuring tools or other useful paraphernalia as will be readily recognized by those of skill in the art.
  • the materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility.
  • the components can be in dissolved, dehydrated, or lyophilized form; they can be provided at room, refrigerated or frozen temperatures.
  • the components are typically contained in suitable packaging material(s).
  • packaging material refers to one or more physical structures used to house the contents of the kit, such as inventive compositions and the like.
  • the packaging material is constructed by well-known methods, preferably to provide a sterile, contaminant-free environment.
  • the packaging materials employed in the kit are those customarily utilized in gene expression assays.
  • a package refers to a suitable solid matrix or material such as glass, plastic, paper, foil, and the like, capable of holding the individual kit components.
  • a package can be a glass vial used to contain suitable quantities of an inventive composition containing primers and probes for RNASET2, TL1A, IFN- ⁇ and/or RNASET2 methylation.
  • the packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components.
  • RNASET2 Loss of RNASET2 in zebrafish results in accumulation of undigested rRNA within lysosomes.
  • the major allele of RNASET2 (i) is a risk for IBD, CD (rs9355610), B1 and both ANCA levels and pos/neg (rs1410925) and (ii) protective for B3 and both ASCA IgA and IgG levels and pos/neg (rs1410925) (Table 2).
  • the major allele rs9355610 is associated with lower levels of RNASET2 mRNA expression in CD small intestine and rectum, EBV transformed B cells and CD3+ PBL from IBD patients.
  • RNASET2 mRNA in CD sigmoid colon The major allele is also associated with RNASET2 mRNA in CD sigmoid colon. Low levels of RNASET2 and increased levels of pANCA are associated with the major allele. Methylation at the RNASET2 locus is inversely correlated with RNASET2 mRNA expression.
  • RNASET2 SNPs Associated with Subclinical Phenotypes of CD PHENO TYPE CHR RSID BP A1 A2 TEST NMISS OR STAT P LOCATION MAF PDM 6 rs3798303 167274358 G A ADD 2093 1.38 3.019 2.54E ⁇ 03 INTRON 0.09853 Iritis 6 rs41269599 167267869 A G ADD 1436 2.806 2.973 2.95E ⁇ 03 INTRON 0.06876 Iritis 6 rs181130555 167277010 D — ADD 1434 2.744 2.932 3.37E ⁇ 03 INTRON 0.07108 Iritis 6 rs3777721 167272065 C G ADD 1436 2.761 2.92 3.50E ⁇ 03 INTRON 0.07084 Iritis 6 rs41269597 167267663 C G ADD 1436 2.711 2.868 4.13E ⁇ 03 INTRON 0.07234 Iritis 6 imm_6
  • RNASET2 Associations with Expression in Small Bowel SNP_rsid gene beta p-value FDR rs72079749 RNASET2 0.151129328 1.66E ⁇ 04 0.371075825 rs66591848 RNASET2 0.154066135 2.27E ⁇ 04 0.42289639 rs1951459 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs4710149 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs933243 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs9355610 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs9356551 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs9366078 RNASET2 0.161888591 2.39E ⁇ 04 0.429556929 rs1819333 RNASET2 0.15479148 3.22E
  • RNASET2 Associations for Expression in Large bowel snp_rsid gene beta t-stat p-value FDR CD Rectum rs683571 RNASET2 ⁇ 0.57902 ⁇ 3.65206 0.00115 0.460077 Rectum rs2031846 RNASET2 ⁇ 0.48099 ⁇ 3.27469 0.002992 0.679028 Rectum rs62436763 RNASET2 ⁇ 0.50894 ⁇ 3.24339 0.003235 0.692932 Rectum rs41463945 RNASET2 ⁇ 0.58473 ⁇ 3.11594 0.004434 0.693762 Sigmoid rs162289 RNASET2 0.147287 2.955434 0.007311 0.619358 Sigmoid rs162291 RNASET2 0.147287 2.955434 0.007311 0.619358 Sigmoid rs162293 RNASET2 0.147287 2.955434 0.007311 0.619358 Sigmoid
  • Allele risk is defined by the Odds ratio (OR). When the A1 allele and an OR of ⁇ 1 is depicted, then the major allele is the risk allele (A2 is risk). If the OR is >1, then the minor allele (A1) is risk allele. Knowing A1 and the Odds Ratio allows you to determine which allele is risk and which is protective.
  • RNAseq The molecular mechanisms of TL1A augmentation of inflammation via enhanced IFN- ⁇ expression were defined using RNAseq.
  • CD4 + T cells were analyzed in untreated conditions or treated with IL12 and IL18 or IL12 and IL18 and TLA1 at the 1 ug scale. On a 10 ng scale, CD4 + T cells were analyzed when treated with IL12 and IL18 and TLA1, either with or without IFN- ⁇ .
  • the RNAseq data prescreen removed all failed probe data, all genes with fewer than 3 samples with FPKM>5. Using this criteria, 8695 genes passed the prescreen and BRB Array Tools were used for class comparison using paired samples.
  • TL1A treatment mediates enhanced expression of IFN- ⁇ , in addition to mediating a decreased expression of RNASET2.
  • the decreased expression of RNASET2 is detected in CD patients: 1) with chronically active disease, 2) with refractive disease requiring surgical intervention, 3) with patients na ⁇ ve to anti-TNF therapy, 4) is associated with OmpC+, ANCA ⁇ serological factors, and 5) is associated with RNASET2 risk SNPs rs9355610, rs1819333, rs2149085.
  • TNFSF15 and the protein it encodes TL1A is associated with IBD and are key mediators of mucosal inflammation.
  • elevated TL1A levels correlate with disease severity and genotype.
  • TL1A mediates marked enhancement of IFN- ⁇ production.
  • TL1A response biomarkers were identified by RNAseq and verified by qPCR in T cells isolated from IBD patients (20 Crohn's [CD], 20 ulcerative colitis [UC]) compared to normal (NL).
  • An additional cohort of samples from NL and IBD patients was used to validate and measure expression/methylation quantitative trait loci (eQTL/mQTL) in the context of GWAS.
  • RNAseq expression clustering differentiated TL1A treated versus non-treated cells.
  • RNASET2 a gene encoding an extracellular T2 RNase, was down-regulated following TL1A treatment.
  • Previous studies associated RNASET2 with susceptibility for CD.
  • RNASET2 expression in CD patients was lower in “severe” vs. mild disease, i.e., multiple disease flare-ups (p ⁇ 0.009), medically refractory (p ⁇ 0.024).
  • siRNA silencing of RNASET2 enhanced TL1A mediated IFN- ⁇ secretion. Without being bound to any particular theory, the inventors believe that down-regulation of RNASET2 is a hallmark of TL1A driven severe CD.
  • RNASET2 expression and DNA methylation were examined in a separate cohort of freshly isolated un-stimulated T cells from NL, CD or UC patients. Methylation at RNASET2 locus inversely correlated with mRNA expression. eQTL of RNASET2 alleles was associated with decreased expression. Significantly enhanced RNASET2 methylation was observed in CD patients with severe disease requiring surgical intervention (Table 14). No correlation was observed in NL or patients with mild disease. Likewise, increased RNASET2 methylation was associated with TNFSF15 risk alleles associated with enhanced TL1A expression (Table 14).
  • RNASET2 epigenetically, the RNASET2 eQTL/mQTL region overlaps with histone H3K4me3 and H3K27ac and DNase HS activation sites. This region co-localizes with transcription factor binding for NF ⁇ B, jun, ATF3 and CEBPD—all of which are up-regulated in response to TL1A treatment.
  • the results identify RNASET2 as a TL1A response gene involved in regulation of IFN- ⁇ production.
  • RNASET2 In CD patients with severe disease there is hyper-methylation and decreased expression of RNASET2, which may be reflective of prior exposure in-vivo to TL1A.
  • RNASET2 serves as a novel potential disease severity biomarker to identify a subset of CD patients most likely to benefit from anti-TL1A therapy.
  • the inventors further analyzed 3 cohorts of patients including 11 UC CD3 + PBT (medically refractive), 43 CD CD3 + PBT (23 were medically refractive and 20 were mild) and 17 normal CD3 + PBT.
  • the samples were run on the Infinium 450 Methylation Array and 11 CD, 12 UC and 4 NL samples were run on the Infinium Expression Array.
  • the inventors demonstrate that RNASET2 expression is decreased following TL1A treatment of CD4 + T cells and that silencing of RNASET2 enhances TL1A mediated IFN- ⁇ secretion.
  • RNASET2 risk allele SNPs rs9355610, rs1819333, rs62436418, rs22236313, rs2769345 RNASET2 risk allele SNPs Refractive disease Normal or Mild disease
  • RNASET2 eQTL/mQTL region overlaps with epigenetic activation sites for 1) histone H3K4me3 and H3K27ac, 2) DNase HS, 3) co-localizes with transcription factor binding for NFkB, jun, ATF3 and CEBPD, all of which are up-regulated in response to TL1A treatment.
  • the enhancer element in primary T memory cells from peripheral blood, the DNAse HS site in CD4 + na ⁇ ve T cells and the eQTL RPS6KA2 monocytes were linked to the rs1819333 allele.
  • TL1A synergizes with IL-12/IL-18 resulting in a rapid (within 6-8 hours) and marked enhancement of IFN- ⁇ expression.
  • RNAseq analysis was used to identify the TL1A response genes regulating IFN- ⁇ expression. Twenty genes were differentially expressed (at least 2-fold) in TL1A activated total CD4+ T cell population. This can be largely due to the fact that IFN- ⁇ secreting T cells constitute only a very small subset (1-3%) of the total CD4+ T cell population ( FIG. 15 ).
  • CD4 T cells from healthy donors were treated with IL12/IL18 and TL1A for 8 hours and then sorted into IFN- ⁇ -secreting and non-secreting subsets ( FIG.
  • a class prediction analysis classifying the IFN- ⁇ -secreting and non-secreting subgroups based on expression levels was performed.
  • the best predictor transcript list consisted of 764 genes with at least two fold differential expression between the IFN- ⁇ secreting subset (p-value ⁇ 0.00005) ( FIG. 17 ).
  • GWAS has identified multiple IBD risk variant SNPs. There was a significant increase in the proportion of transcripts located within 0.5 MB from an IBD risk SNPs (14% vs.
  • RNASET2 a member of the Rh/T2/S family of ribonuclease, was the only IBD risk associated gene displaying a greater than 5 fold TL1A mediated down-regulation in expression. RNASET2 has been identified by GWAS as a potential IBD risk gene. As the functional role of RNASET2 in IBD pathogenesis was unknown, the regulation of RNASET2 expression in IBD was examined.
  • RNASET2 is a ‘class predictor’ gene, differential expression can be detected in total CD4+ T cells. Resting or IL12/IL18 treated CD4+ T cells from CD, UC patients or NL controls were isolated and RNASET2 levels were compared in the presence or absence of TL1A for 8 hours. As seen in FIG. 7 , in contrast to what we had been observed in cells from NL donors, IBD patients did not display a TL1A mediated decrease in RNASET2 expression levels. Rather decreased expression levels of RNASET2 were associated in CD patients with “severe” compared to mild disease course.
  • RNASET2 expression was significantly lower in cells isolated from CD patients exhibiting multiple disease flares per year (p ⁇ 0.001) ( FIGS. 8A and 8C ) and likewise decreased RNASET2 expression was detected in CD patients who were medically refractive requiring surgical intervention for disease management (p ⁇ 0.024) ( FIG. 8B ). A similar trend was seen in UC patients.
  • Gene expression quantitative trait was performed to characterize the functional correlation between RNASET2 gene variation and the gene transcript expression level.
  • the data indicates a pathway whereby down-regulation of RNASET2 alters IFN- ⁇ expression.
  • the functional role of RNASET2 in regulation of IFN- ⁇ expression was confirmed using siRNA mediated silencing.
  • CD4+ T cells were transfected with siRNA targeting RNASET2 mRNA or control siRNA and then treated with IL12/IL18 and TL1A.
  • RNASET2 mRNA itself displayed a 60-70% inhibition by RNASET2 siRNA ( FIG. 21A ).
  • a significant enhancement (>1.5 fold) in IFN- ⁇ expression was seen in cells transfected with RNASET2 siRNA compared to control scrambled siRNA ( FIG. 21B ).
  • RNASET2 expression was examined in a separate cohort of freshly isolated unstimulated T cells from NL, CD and UC patients. Since DNA methylation impacts upon gene expression and since most disease-risk genetic polymorphisms map outside of the transcribed exome in regions subject to epigenetic modification, the DNA methylation status of RNASET2 was examined as well. In unstimulated T cells from CD and UC patients an inverse correlation was observed between IFN ⁇ expression levels and RNASET2 ( FIG. 22 ).
  • RNASET2 gene variation was confirmed in unstimulated peripheral T cells from IBD patients with refractory disease ( FIG. 11 ), with decreased expression correlating with the RNASET2 risk allele variant SNP. Moreover, a similar eQTL was observed for mRNA extracted from tissue obtained from surgical resection of the small bowel using gene expression microarray ( FIG. 12 ). The correlation between RNASET2 gene variation and methylation, mQTL was also examined and a significant mQTL was observed with an increase in methylation in IBD patients with refractory disease ( FIGS. 13A-13D ). In contrast, no mQTL was detected in cells isolated from patients with mild disease or NL subjects.
  • RNASET2 down-modulation of RNASET2 is a component of TL1A mediated enhancement of IFN- ⁇ expression. Additionally, epigenetic modulation of RNASET2 and reduced gene expression in IBD patients with a known IBD risk variant SNP is associated with a more severe course of disease.
  • RNA-seq RNA sequencing
  • qPCR quantitative polymerase chain reaction
  • IFN- ⁇ interferon gamma
  • ELISA Enzyme-linked immunosorbent assay
  • siRNA small interfering RNA
  • Findings were validated using additional samples of unstimulated T cells from NL and IBD patients or small bowel (SB) surgical resection and analyzed for expression quantitative trait loci (eQTL) and methylation quantitative trait loci (mQTL) based on genotyping and clinical data.
  • eQTL quantitative trait loci
  • mQTL methylation quantitative trait loci
  • TF binding sites Screening for predicted motif disruption of transcription factor (TF) binding sites identified candidate regulatory SNPs. Proteomic analysis and measurement of cytokine secretion were used to determine effect of RNASET2-directed small interfering RNA (siRNA) on protein expression. Cell aggregation was measured by flow cytometry.
  • siRNA small interfering RNA
  • IBD patients were defined as “refractory” if surgical intervention was required for disease management following failure of medical therapy. IBD patients were defined as “mild” if they had no prior surgeries and no active disease at time of sample collection. CD patients exhibiting one or more disease flares per year were defined to have “severe disease” compared to patients with no disease flares per year. Clinical characteristics were prospectively collected from 564 CD patients who had undergone surgical resection.
  • PBMC Peripheral blood mononuclear cells
  • CD3 + T cells PBT
  • CD3-immunomagnetic beads Miltenyi Biotech, Auburn, Calif.
  • CD4 + T cells were isolated using negative selection by depletion with magnetic beads (Stemcell Technologies, Vancouver, BC, Canada) and were at least 95% pure.
  • DNA samples from CD3 + T cells were bisulfite converted using the Zymo EZ DNA Methylation kit (Zymo Research) with an input of 1 ⁇ g.
  • the assay was carried out as per the Illumina Infinium Methylation instructions, using the Infinium HumanMethylation450 BeadChip Kit (Illumina Inc., San Diego, Calif.). Data were visualized and normalized using the GenomeStudio software.
  • the methylation ⁇ values were recalculated as the ratio of (methylated probe signal)/(total signal).
  • IFN- ⁇ was measured by an amplified ELISA.
  • Greiner Bio-One (Longwood, Fla.) ELISA plates were coated overnight with 100 ⁇ l of 5 ⁇ g/ml monoclonal anti-IFN- ⁇ (BD Biosciences, Woburn, Mass.). Samples and standards were added for 24 h followed by addition of 100 ⁇ l of 2.5 ⁇ g/ml polyclonal biotinylated rabbit anti-IFN- ⁇ (BD Biosciences) for 2 h. This was followed by addition of 100 ⁇ l of 1/1000 diluted alkaline phosphatase-conjugated streptavidin (Jackson ImmunoResearch Laboratories, West Grove, Pa.) for 2 h.
  • Substrate 0.2 mM NADP (Sigma-Aldrich, St. Louis, Mo.) was added for 30 min followed by addition of amplifier (3% 2-propanol, 1 mM iodonitrotetrazolium violet, 75 ⁇ g/ml alcohol dehydrogenase, and 50 ⁇ g/ml diaphorase; Sigma-Aldrich) for 30 min. Plates were read at 490 nm using an E max plate reader (Molecular Devices, Sunnyvale, Calif.).
  • RNASET2 siRNA or control siRNA 600 V, for 9 pulses of 500 ⁇ sec, with 100 ⁇ sec between pulses
  • 4 mm gap width cuvettes in a BTX Electro Square Porator ECM 830 (Genetronics, Inc., San Diego, Calif.). Sequences used in siRNA inhibition are depicted in Table 16.
  • siRNA sequences SEQ Sequence Name Sequence ID NO RNASET2 siRNA-sequence forward 5′-GCAAGAGAAAUUCACAAACUGCAGC-3′ 7 RNASET2 siRNA-sequence reverse 5′-GCUGCAGUUUGUGAAUUUCUCUUGCUU-3′ 8 Control siRNA-sequence forward 5′-CUUCCUCUCUUUCUCUCCCUUGUGA-3′ 9 Control siRNA-sequence reverse 5′-UCACAAGGGAGAGAAAGAGAGGAAGGA-3′ 10
  • Tandem mass tagging (TMT)-based quantitative proteomics analysis was conducted as described (Qu et al., Sci Rep 2016; 6:32007). For each sample, 50 ⁇ g proteins were digested in parallel into tryptic peptides using filter-aided sample preparation (FASP) (Wisniewski et al., Nat. Methods 2009; 6:359-62). Peptides derived from eight samples and a pooled internal standard were labeled with a set of TMT10plex reagents (Thermo Scientific), mixed, desalted, separated into 24 fractions by high-pH liquid chromatography, and concatenated into 8 fractions.
  • FASP filter-aided sample preparation
  • Fractionated peptides were resolved on a 50 cm EASY-Spray analytical column, and analyzed by an LTQ Orbitrap Elite mass spectrometer (Thermo Scientific) in the data-dependent acquisition mode, using the higher-energy collisional dissociation (HCD) method for tandem mass spectrometry.
  • HCD collisional dissociation
  • Acquired raw data were searched against the human Uniprot database (released on Oct. 17, 2015, containing 20,982 sequences) with Proteome Discoverer (v2.1), using the SEQUEST algorithm.
  • a stringent 1% false discovery rate was set to filter peptide and protein identifications. Peptides with >30% precursor ion interference were excluded from protein quantification.
  • IFN- ⁇ -secreting CD4 + T cells was isolated by flow cytometry following activation of cells with recombinant human IL-12 (500 pg/ml, R&D Systems, Minneapolis, Minn.) and IL-18 (50 ng/ml, R&D Systems) and TL1A (100 ng/ml, Fitzgerald Industries International, Acton, Mass.) for 8 h.
  • IFN- ⁇ -secreting CD4 + T cells were detected using an IFN- ⁇ secretion assay cell enrichment and detection kit (Miltenyi Biotec, San Diego, Calif.). Cells were sorted on a FACS Aria II (BD Biosciences, San Jose, Calif.).
  • Intracellular IFN- ⁇ production and analysis of cellular aggregation was conducted essentially as described (Dezorella et al., Cytometry B Clin Cytom 2016:90:257-66) Briefly, cells were either rested or stimulated for 24 h with IL12/IL18 and TL1A and Berfeldin A (10 ug/ml) was added for the last 4 h. Cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 and 0.2% saponin and stained for intracellular IFN- ⁇ (brilliant violet 421-IFN- ⁇ , eBioscience) or isotype control. Samples were washed and stained for cellular aggregation (propidium iodide).
  • Genotype data was obtained for Caucasian subjects using Illumina HumanImmuno BeadChip array. Markers were excluded based on: test of Hardy-Weinberg Equilibrium with significance threshold of p ⁇ 10 ⁇ 3 ; if genotyping rate was ⁇ 100% (for eQTL and mQTL associations) or ⁇ 98% (for GWAS) and if minor allele frequency was ⁇ 5%. Identity-by-descent was used to exclude related individuals (Pi-hat scores >0.25) using PLINK. ADMIXTURE was used to perform ethnicity analysis to get ethnicity proportion estimation for individuals. An individual with Caucasian proportion ⁇ 0.75 was classified as Caucasian.
  • CD cases and 33,938 non-IBD controls genotyped using ImmunoChip were included in the analysis after samples with >5% missing data, samples of non-European ancestry from population stratification or with abnormal mean intensity values, and SNPs with >2% missing data or HWE p-value ⁇ 10 ⁇ 10 in controls were removed.
  • 13511 have disease behavior information collected based on Montreal classification as reported previously (Cleynen et al., Lancet 2016; 387:P156-67) (described as B1, non-stricturing, non-penetrating, B2, stricturing and B3, penetrating diseases).
  • Single channel microarray expression data extracted using Agilent feature extraction software were received from Genome Technology Access Center at Washington University, St. Louis.
  • Raw expression data available in technical duplicates were normalized using LIMMA package implemented in R version 3.2.2.
  • the expression data preprocessing included background correction of the expression data, followed by log 2-transformation and quantile-normalization.
  • Pathway analysis was accomplished through the use of Qiagen's Ingenuity® Pathway Analysis (IPA®, Qiagen, Redwood City, www.qiagen.com/ingenuity) and The Database for Annotation, Visualization and Integrated Discovery (DAVID, http://david.abcc.ncifcrf.gov).
  • IPA® Qiagen's Ingenuity® Pathway Analysis
  • DAVID The Database for Annotation, Visualization and Integrated Discovery
  • Modeling, data analysis, and data mining were performed using the BRB array tools (brb.nci.nih.gov/BRB-ArrayTools) and R-program (version 2.2.2; www.r-project.org).
  • Cluster analysis was performed using Cluster 3.0 and Java Treeview 1.1.6r4. Tests for statistical significance were determined using JMP Statistical Software (Cary, N.C.).
  • RNASET2 an IBD susceptibility gene
  • IBD susceptibility gene an IBD susceptibility gene
  • RNASET2 disease-risk variants for decreased expression (eQTL) in peripheral and mucosal tissues and DNA hypermethylation (mQTL) from CD patients with medically refractory, but not mild disease. Additionally, RNASET2 disease-risk variants were associated with an increase in development of stricturing/penetrating disease behavior. Furthermore, RNASET2 disease-risk variants were associated with a complicated/resistant CD phenotype defined in part by therapeutic drug failure, ANCA sero-positivity, increased length of intestinal resection, shorter time to reoperation and post-operative endoscopy with a high (>2) Rutgeerts score.
  • RNASET2 disease-risk variants identified rs2149092 with predicted disruption of a consensus ETS-TF binding site located within a potential enhancer region, providing insight into RNASET2 cis-regulatory elements.
  • RNASET2 correlated with expression of multiple ETS-transcription factors.
  • siRNA silencing of RNASET2 resulted in enhanced IFN- ⁇ , increased ICAM1 and concomitant T cell aggregation while anti-LFA1 blocking of aggregation suppressed IFN- ⁇ secretion.
  • a key IBD proinflammatory cytokine CD4 + T cells were treated with TL1A, sorted into IFN- ⁇ -secreting and non-secreting subsets and analyzed by RNA-seq ( FIG. 15 and FIG. 27 ).
  • Unsupervised hierarchical clustering of the set of expressed genes clearly distinguished TL1A-mediated IFN- ⁇ -secreting and non-secreting groups ( FIG. 16 ). Seven hundred and sixty-four “predictor” genes with at least two-fold differential expression between the IFN- ⁇ secreting/non-secreting subsets (p value ⁇ 1 ⁇ 10 ⁇ 5 ) ( FIG. 17 ) were identified.
  • RNASET2 was the only IBD risk associated gene with greater than 5-fold down regulation in the IFN- ⁇ secreting CD4 + subset.
  • RNASET2 is the only human member of the Rh/T2/S family of ribonucleases and its expression is decreased in ovarian cancer, melanoma and non-Hodgkin lymphoma. Considering the key role for IFN- ⁇ in pathogenesis of Crohn's disease/IBD, without being bound to any particular theory, these data collectively suggest that down regulation of RNASET2 identifies TL1A mediated ‘severe’ CD. RNASET2 expression was examined in freshly-isolated, unstimulated peripheral CD3 + T cells from a separate cohort of NL, CD and UC patients.
  • RNA-seq analysis demonstrated there was an inverse correlation between TNFSF15 expression levels and RNASET2 in peripheral T cells from two independent cohorts comprised of a combined 138 CD patients ( FIGS. 24A and 24D ). These results were consistent even when each cohort was analyzed separately ( FIGS. 24B and 24C ). Moreover, there was a significant negative correlation between expression and methylation ( FIG. 23 ), mainly within 50 kb upstream and downstream from the transcriptional start site (TSS) ( FIG. 26 ).
  • RNASET2 Disease-Risk Alleles are Associated with Decreased RNASET2 Expression and Increased DNA Methylation in CD Patients with Refractory Disease
  • RNASET2 gene variation was performed to characterize the functional correlation between RNASET2 gene variation and the gene transcript expression level.
  • the functional correlation between RNASET2 IBD-risk genotypes and the gene transcript expression levels were established in unstimulated peripheral CD3 + T cells isolated from IBD patients with refractory disease.
  • RNASET2 expression in T cells from subjects carrying the RNASET2 risk alleles rs2149085, rs1819333, and rs9355610 FIG. 11 .
  • These findings were confirmed with significant eQTL observed for mRNA extracted from uninflamed small bowel tissue obtained from CD subjects at surgical resection ( FIG. 12 ).
  • the correlation between RNASET2 gene variation and methylation, mQTL was also examined. A significant mQTL was observed with an increase in methylation in CD patients with refractory disease ( FIGS. 13A and 13C ). In contrast, no mQTL was detected in cells isolated from CD patients with mild disease or NL subjects ( FIGS. 13B and 13D ).
  • RNASET2 In IBD pathogenesis, regulation of RNASET2 expression in CD4 + T cells isolated from IBD patients was examined and compared to normal (NL) donors in the presence or absence of TL1A. As seen in FIG. 7 , NL donors, but not IBD patients, exhibited a TL1A-mediated decrease in RNASET2 expression levels. Instead, decreased expression levels of RNASET2 were found in CD patients with more severe disease (exhibiting one or more disease flares per year) compared to patients with no yearly disease flares ( FIGS. 8A and 8C ).
  • RNASET2 Disease Risk Alleles are Associated with Complicated and Resistant Disease Behavior
  • RNASET2 disease risk variant SNPs were associated with a complicated stricturing/penetrating phenotype (Montreal classification B1 vs. B2 and B3), (Table 18).
  • RNASET2 disease risk variant SNPs were associated with therapeutic failure of thiopurine or anti-TNF therapy, ANCA sero-positivity (a marker associated with lack of response to anti-TNF therapy), and an increased length of intestinal resection characteristic attributed to overall disease severity (Table 18 and FIGS. 38-39 ). No association was observed for therapeutic failure on steroids or sulfasalazine. Moreover, patients with RNASET2 disease risk variant SNPs who required multiple resections for disease management exhibited a shorter time to reoperation ( FIG. 32 ).
  • RNASET2 risk SNPs were associated with a more severe disease recurrence.
  • Post-operative endoscopies revealed an association of RNASET2 risk SNPs in patients classified with a high Rutgeerts score (>2) who were not receiving postoperative prophylaxis (Table 18). No association was observed for clinical recurrence.
  • Decreased expression of RNASET2 was also associated with a penetrating disease phenotype ( FIG. 31 ) and ASCA sero-positivity ( FIG. 30 ). Without being bound by any particular theory, this data supports an association of RNASET2 disease risk SNPs with clinical parameters suggestive of complicated and resistant disease behavior.
  • RNASET2 Variant in LD with Disease-Tagging SNP Disrupts ETS Transcription Factor Binding Motif
  • RNASET2 risk variants demonstrate significant overlap between more than a hundred CD RNASET2 risk variants, many in linkage disequilibrium, associated with eQTL and mQTL creating difficulty in determining functionality/causality. Since the majority of RNASET2 risk variants associated with CD are located in non-coding regions, it is likely that these SNPs alter expression through modulation of regulatory functions. Furthermore, without being bound to any particular theory, studies suggest that SNPs associated with disease often exist within active enhancer regions of cell types relevant to disease and can disrupt TF binding motifs. REMC data demonstrate that the RNASET2 locus is marked in primary T cells, compared to other tissues, by putative active enhancer histone modifications and active gene expression ( FIG. 40 ).
  • RNASET2 Enhances IFN- ⁇ Secretion Via Upregulation of ICAM1 Expression and Homotypic T Cell Aggregation
  • RNASET2 The functional role of RNASET2 in regulation of IFN- ⁇ secretion was tested using siRNA silencing.
  • CD4 + T cells transfected with siRNA targeting RNASET2 mRNA followed by stimulation with TL1A.
  • Cells transfected with siRNA targeting RNASET2 displayed a 60-70% inhibition of RNASET2 expression ( FIGS. 21A and 37A ), and a parallel significant enhancement (>1.5 fold) in TL1A mediated IFN- ⁇ secretion was seen compared to control siRNA ( FIG. 21B and FIG. 37B ).
  • FIGS. 21A and 37A show that down regulation of RNASET2 modulates IFN- ⁇ expression.
  • Candidate targets were selected on the basis of exhibiting both modulation of expression following siRNA silencing of RNASET2 as well as TL1A-mediated differential expression when comparing IFN ⁇ secreting and non-secreting T cells (data from RNAseq analysis).
  • ICAM1 One of the proteins that was up-regulated in response to RNASET2 silencing and in the IFN ⁇ secreting compared to non-secreting T cells, was ICAM1 ( FIG. 42 ). ICAM1 was recently identified as an IBD susceptibility locus, with up-regulated gene expression associated with the disease-risk variant.
  • ICAM1 is a transmembrane adhesion protein commonly expressed by vascular endothelium and leukocytes. Binding of ICAM to the LFA1 receptor on T cells facilitates and stabilizes cell-cell interactions. Studies have demonstrated increased ICAM1 expression on activated T cells and proposed a role for ICAM1-LFA1 binding in inducing homotypic T cell aggregation and subsequent T cells differentiation. To examine the effect of cell-cell contact on TL1A mediated IFN- ⁇ secretion, cells were incubated in flat bottom and conical bottom microwells. A greater than 3 fold increase in IFN- ⁇ production was consistently observed when cells were incubated in close cell-cell conical geometry (data not shown).
  • T cells were stimulated in the presence or absence of TL1A and then stained with an antibody for intracellular IFN- ⁇ ( FIGS. 37C and 37D , left panels) and for cellular aggregation using propidium iodide (PI) ( FIGS. 37C and 37D , upper and lower right panels).
  • the PI-labeled peaks correspond to number of cells per event allowing for identifying single cells versus cellular aggregates.
  • the first peak in each histogram corresponds to single cell events (black brackets) and the successive peaks, to multicellular aggregates (gray brackets).
  • TL1A-mediated downregulation of RNASET2 and concomitant enhancement of ICAM1 expression promotes homotypic T cell aggregation and augmentation of IFN- ⁇ production. It is noted that an increase in expression of ICAM1 was associated in CD with ASCA sero-positivity and pre-op therapeutic failure of anti-TNF and thiopurine ( FIG. 43 ), clinical parameters associated with decreased RNASET2 and disease activity.
  • RNASET2 and TNFSF15 have been identified among the 201 GWAS IBD susceptibility loci.
  • RNASET2 risk SNPs were associated with clinical parameters suggestive of a complicated and resistant disease behavior.
  • response to therapeutics following surgery and recurrence of disease were associated with RNASET2 risk SNPs.
  • IRF4 is an IBD susceptibility SNP that is lymphocyte specific and is essential for the differentiation of Th1, Th2, Th9, Th17 and T reg subsets.
  • ELF-1 is a CD susceptibility SNP in the Japanese population. It is an ETS family transcription factor that is expressed in lymphoid cells, acts as both an enhancer and a repressor of expression and is involved in IL2 and IL23 signaling.
  • PU.1 is also an ETS family transcription factor and is essential for early stages of T cell development. It down regulates ⁇ T Cells which are found in the mucosa and plays a role in innate immunity and when expressed in T H 9 cells, these cells drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells.
  • RNASET2 expression is decreased following TL1A treatment in IFN- ⁇ secreting CD4 + T cells and that silencing of RNASET2 enhances TL1A mediated IFN- ⁇ secretion.
  • Clinical correlates have also been identified for RNASET2 disease associated SNPs, which include, but are not limited to therapeutic failure of thiopurine therapy, therapeutic failure of anti-TNF therapy, ANCA Sero-positivity, B2/B3 versus B1 (structuring/penetrating vs non-penetrating/non-stricturing) disease, an increase in length of intestinal resection, decreased time to second surgery and endoscopic recurrence of disease with high Rutgeerts score.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention describes methods of diagnosing inflammatory bowel disease, including but not limited to Crohn's Disease (CD), Ulcerative Colitis (UC), and/or Medically Refractive Ulcerative Colitis (MR-UC), using RNA-SET2, TL1A and/or IFN-Υ. The invention further provides a process for patient identification and/or stratification.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a U.S. National Phase of International Application No. PCT/US2017/023082 filed Mar. 17, 2017, which claims the benefit of U.S. Provisional Ser. Nos. 62/457,048 filed Feb. 9, 2017 and 62/309,817 filed Mar. 17, 2016, all of which are incorporated herein in their entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This invention was made with government support under Grant Nos. DK043211, DK046763, DK062413, HS021747, AI067068, DE023798, DK084554, RR033176 and TR000124 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • FIELD OF INVENTION
  • The invention relates to inflammatory bowel disease and RNASET2 as a biomarker for disease severity and targeting anti-TL1A therapy.
  • BACKGROUND
  • All publications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The following description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed invention, or that any publication specifically or implicitly referenced is prior art.
  • Inflammatory bowel disease (IBD) has two common forms, Crohn's disease (CD) and ulcerative colitis (UC), which are chronic, relapsing inflammatory disorders of the gastrointestinal tract. Genetic factors play an important role in IBD pathogenesis, as evidenced by the increased rates of IBD in Ashkenazi Jews, familial aggregation of IBD, and increased concordance for IBD in monozygotic compared to dizygotic twin pairs (S. Vermeire, P. Rutgeerts, Genes Immun 6, 637 (2005)). Moreover, genetic analyses have linked IBD to specific genetic variants. CD and UC are thought to be related disorders that share some genetic susceptibility loci but differ at others.
  • IBD is generally believed to be triggered in genetically susceptible individuals by an inappropriate immune response to the commensal flora. The high clinical heterogeneity and genetic complexity of CD and UC suggest that the underlying biological pathways driving disease almost certainly differ in subgroups of patients. Thus, the development of early and targeted therapeutics requires subgroup stratification and prognostic biomarker identification, particularly in predicting an overall mild, compared to severe, disease course. Although 201 IBD susceptibility loci have been identified, little is known regarding their functional significance. Genetic variation in TNFSF15 is associated with CD in multiple populations, and the protein it encodes, TL1A, is a key mediator of mucosal inflammation. TL1A expression is up-regulated in inflamed regions of the intestine in both CD and UC. In IBD patients, elevated TL1A levels correlate with TNFSF15 genotype and disease severity. CD patients with elevated serum/tissue levels of TL1A have increased risk of developing fibrosis/stricturing disease behavior. In vitro, TL1A synergizes with interleukin 12 (IL-12) and interleukin 18 (IL-18) (12/18), leading to rapid enhancement of IFN-γ production, another key mediator of mucosal inflammation.
  • Therefore, there remains a need in the art for methods of diagnosing and identifying patients for treatment with IBD, CD, UC and/or MR-UC.
  • BRIEF DESCRIPTION OF THE FIGURES
  • Exemplary embodiments are illustrated in referenced figures. It is intended that the embodiments and figures disclosed herein are to be considered illustrative rather than restrictive.
  • FIG. 1 depicts RNASET2 eQTL microarray in uninvolved small intestine, in accordance with various embodiments of the invention.
  • FIG. 2 shows that the RNASET2 major allele is associated with decreased expression of RNASET2 in sigmoid colon and rectum of CD patients, in accordance with various embodiments of the invention.
  • FIG. 3 shows that the RNASET2 major allele is associated with decreased expression of RNASET2 in sigmoid colon and rectum of UC patients, in accordance with various embodiments of the invention.
  • FIG. 4 shows that the RNASET2 risk allele is associated with decreased expression of RNASET2 in inflamed large bowel in CD and UC patients, in accordance with various embodiments of the invention. Similar results were observed for rs1819333.
  • FIG. 5 shows that the RNASET2 major allele is associated with decreased RNASET2 expression in small bowel from CD surgeries, in accordance with various embodiments of the invention.
  • FIG. 6 depicts RNASET2 eQTL in EBV Transformed B Cell Lines, in accordance with various embodiments of the invention. The major allele is associated with lower levels of RNASET2 mRNA expression in EBV transformed B cell lines.
  • FIG. 7 depicts RNASET2 expression following IL-12, IL-18, and/or TL1A and treatment, in accordance with various embodiments of the invention.
  • FIGS. 8A-8C depict RNASET2 expression in CD patients, in accordance with various embodiments of the invention. A) RNASET2 expression in patients with none or 1 multiple disease flares per year. B) RNASET2 expression following TL1A treatment in patients who were medically refractive requiring surgical intervention for disease management or no surgery. C) RNASET2 expression based upon disease flares per year in 32 CD patients, encompassing additional data samples.
  • FIG. 9 shows a decreased expression of RNASET2 in IBD patients with RNASET2 risk alleles, in accordance with various embodiments of the invention.
  • FIG. 10 depicts RNASET2 methylation versus GWAS p values in CD and UC patients, in accordance with various embodiments of the invention.
  • FIG. 11 depicts the eQTL of RNASET2 in refractory IBD, in accordance with various embodiments of the invention. RNASET2 SNPs (rs2149085, rs1819333 and rs9355610) from CD3+ peripheral T cells from 11 CD and 10 UC patients requiring surgical intervention for disease management, using an Illumina expression array.
  • FIG. 12 depicts the eQTL of RNASET2 in CD small bowel (ileal) surgical resection of 85 CD patients using an Agilent expression array, in accordance with various embodiments of the invention.
  • FIGS. 13A-13D depict the mQTL of RNASET2 in refractory IBD, in accordance with various embodiments of the invention. A) mQTL of RNASET2 in refractory IBD and B) normal or mild disease patients. C) mQTL (cg25258033) of CD3+ peripheral T cells from 20 CD patients with refractory disease requiring surgical intervention for disease management and D) 16 patients who were responsive to IBD therapeutics and 9 normal controls, encompassing additional data samples.
  • FIGS. 14A-14B depict the mapping of eQTL and mQTL across RNASET2 in patients with refractory or mild disease, in accordance with various embodiments of the invention. A) eQTL and mQTL across RNASET2 in patients with refractory or mild disease. B) eQTL and mQTL calculated using CD3+ T cells from both the periphery and mucosal compartments from patients with refractory or mild disease (including normal patients), encompassing additional data samples.
  • FIG. 15 depicts sorting and IFNγ expression of CD4+ T cells stimulated with IL-12, IL-18 and/or TL1A, in accordance with various embodiments of the invention. Histograms of side scatter vs. IFN-γ for CD4+T cells stimulated with recombinant human IL-12 (500 pg/ml) and IL-18 (50 ng/ml) and TL1A (100 ng/ml) for 8 h from 4 donor (D1-4).
  • FIG. 16 depicts a dendrogram of hierarchical clustering using centered correlation and average linkage, in accordance with various embodiments of the invention.
  • FIG. 17 depicts a class prediction analysis classifying the IFNγ-secreting and non-secreting subgroups based on expression level, in accordance with various embodiments of the invention. Heatmap of 764 predictor genes.
  • FIG. 18 depicts a proportion of genes differentially expressed that was increased in GWAS versus other regions, in accordance with various embodiments of the invention.
  • FIG. 19 depicts 183 transcribed IBD associated SNP regions in the T cells, in accordance with various embodiments of the invention.
  • FIG. 20 depicts a volcano plot of the class predictor GWAS transcripts of IBD risk predictor genes, in accordance with various embodiments of the invention.
  • FIGS. 21A-21B show that silencing RNASET2 enhances IFN-γ secretion, in accordance with various embodiments of the invention. A) Inhibition of RNASET2 by RNASET2 siRNA. B) Effect of RNASET2 silencing on IFN-γ secretion. Enhanced IFN-γ expression in cellos transfected with RNASET2 siRNA compared to control scrambled siRNA.
  • FIG. 22 demonstrates the inverse correlation of IFN-γ and RNASET2 expression in accordance with various embodiments of the invention.
  • FIG. 23 demonstrates a negative correlation of RNASET2 methylation and expression in CD3+ T cells (cg25258033, located 1.4 kb within the first intron) in 21 IBD patients, in accordance with various embodiments of the invention.
  • FIGS. 24A-24D depict correlation of RNASET2 and TNFSF15 expression in CD3+ T cells from patients with refractory disease requiring surgical intervention for disease management, using RNA-seq, in accordance with various embodiments of the invention. A) Correlation of RNASET2 versus TL1A in refractory CD. B) Correlation of RNASET2 and TNFSF15 expression in CD3+ peripheral T cells from 38 CD patients, C) depicts data from 100 CD patients and D) depicts combined data from 138 patients.
  • FIG. 25 depicts the correlation expression of RNASET2 versus A) PU.1 and B) ELF1, in accordance with various embodiments of the invention. The risk SNP rs2149092 C/T (SEQ ID NO: 2) abolishes the IRF4, PU.1, and ELF-1 binding site. C—non-risk and T=risk allele.
  • FIG. 26 depicts correlation of expression and methylation located within 100 kb of the RNASET2 transcriptional start site in 21 IBD patients, in accordance with various embodiments of the invention.
  • FIG. 27 depicts IFN-γ expression of IFN-γ producing and non-producing levels in CD4+ T cells, in accordance with various embodiments of the invention.
  • FIG. 28 depicts the correlation of GWAS p values with eQTL p values over the RNASET2 locus, in accordance with various embodiments of the invention. GWAS p values are based upon data from 18729 CD and 34897 controls. eQTL p values are based upon genotyping and RNA-seq based expression of RNASET2 for 71 CD patients with refractory disease, requiring surgical intervention for disease management.
  • FIG. 29 depicts the effect of RNASET2 silencing on IFN-γ secretion, in accordance with various embodiments of the invention. Inhibition of RNASET2 expression by RNASET2-specific siRNA was greater than 50% in all experiments.
  • FIG. 30 depicts an association of decreased expression of RNASET2 with ASCA Sero-positivity, in accordance with various embodiments of the invention.
  • FIG. 31 depicts an association of a decreased expression of RNASET2 with penetrating disease, in accordance with various embodiments of the invention. Expression of RNASET2 by RNA-seq for 71 CD patients based upon Montreal disease classification (B1, B2, and B3).
  • FIG. 32 depicts that patients with RNASET2 disease associated SNPs exhibited a shorter time to reoperation, in accordance with various embodiments of the invention. Time between surgeries based upon carriage for IBD risk SNP rs9355610 (SEQ ID NO: 3) for 154 CD patients who underwent multiple surgeries.
  • FIG. 33 depicts that the rs2149092 SNP (SEQ ID NO: 2) Alters DNA Shape.
  • FIG. 34 depicts the correlation of RNASET2 versus Ets1 expression, in accordance with various embodiments of the invention.
  • FIG. 35 depicts that the rs2149092 SNP (SEQ ID NO: 2) distorts DNA shape at the Ets1 binding site.
  • FIGS. 36A-36C depict identification of potential regulatory function of RNASET2 disease associated variant rs2149092 (SEQ ID NO: 2) the prospective regulatory role of RNASET2 variant rs2149092 (C-non-risk allele/T-risk allele), in accordance with various embodiments of the invention. A) Predicted disruption of rs2149092 C to T variation in the binding motifs for ETS and IRF4 transcription factors. Central ETS in the variant motif is underlined. B) CHIP-seq and histone modification profiles for ETS1, IRF4 and SPI1 transcription factor binding and histone H3K4me1 and H3K4ac aligned with the genomic sequence surrounding rs2149092 variant. C) Correlation of expression of RNASET2 and multiple ETS and JUN transcription factors in CD3+ peripheral T cells from 108 CD patients requiring surgical intervention for disease management, using RNA-seq.
  • FIGS. 37A-37G depict the effect of RNASET2 silencing on IFN-γ secretion and cellular aggregation, in accordance with various embodiments of the invention. A) Silencing of RNASET2 expression by RNASET2 or control (NC) siRNA. B) Effect of RNASET2 silencing on IFN-γ secretion. Panels A and B are representative of 6 out of 7 experiments (FIG. 29) with similar results. C) CD4+ T cells were either not treated (UT) or D) stimulated with TL1A for 24 hours. Intracellular IFN-γ staining and cellular aggregations were measured by flow cytometry. Cells were gated on IFN-γ secreting and non-secreting populations (left panels) and then using propidium iodide (PI) analyzed for single and aggregate cell fractions (histograms, right panels). The first peak in each histogram corresponds to single cells (black bracket) and the remaining peaks to cellular aggregates (gray bracket). Representative of 4 experiments. E) Proportion of single cells and cellular aggregates in IFN-γ secreting (IFN-γ+) and non-secreting (IFN-γ−) populations following TL1A stimulation. F) Fold increase in number of IFN-γ secreting cells (Average of 4 experiments). G) CD4+ T cells were pretreated with control IgG or LFA1 blocking Ab (aLFA1) prior to TL1A stimulation. Overall p value for LFA1 mediated blocking of IFN-γ secretion, measured by ELISA, was 0.047.
  • FIG. 38 depicts in accordance with various embodiments of the invention, the association of RNASET2 disease risk variant SNPs at the time of surgery with therapeutic failure of thiopurine or anti-TNF therapy, ANCA sero-positivity and an increased length of intestinal resection (data summarized in Table 18).
  • FIG. 39 depicts in accordance with various embodiments of the invention, the association of RNASET2 disease risk variant SNPs with disease recurrence in 38 patients who were not receiving postoperative prophylaxis. Post-operative endoscopies were performed and classified by Rutgeerts score. (data summarized in Table 18).
  • FIG. 40 depicts tissue-specific functional annotation of RNASET2 locus. Heatmap of H3K4me3, H3K4me1 and RNAseq data from REMC.
  • FIG. 41 depicts the correlation of RNASET2 expression and multiple ETS and IRF4 transcription factors in CD3+peripheral T cells from 108 CD patients with refractory disease, using RNA-seq.
  • FIG. 42 depicts in accordance with various embodiments of the invention, a heat-map illustrating protein expression of indicated genes in CD4+T cells relative to untreated cells following silencing with control (NC) or RNASET2 siRNA. Results are from 4 healthy donors. The right column depicts differential gene expression in IFN-γ secreting compared to non-secreting CD4+T cells.
  • FIG. 43 depicts in accordance with various embodiments of the invention, clinical disease parameters associated with level of ICAM1 expression (measured by RNA-seq) for 71 CD patients based upon IgG ASCA sero-positivity (left panel) or pre-op therapeutic failure of anti-TNF (middle panel) or thiopurine (right panel).
  • DESCRIPTION OF THE INVENTION
  • All references cited herein are incorporated by reference in their entirety as though fully set forth. The sequences related to RNASET2 are also incorporated by reference in their entirety as though fully set forth via the rs number disclosed. Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 3rd ed., Revised, J. Wiley & Sons (New York, N.Y. 2006); and Sambrook et al., Molecular Cloning: A Laboratory Manual 4th ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y. 2012), provide one skilled in the art with a general guide to many of the terms used in the present application. For references on how to prepare antibodies, see D. Lane, Antibodies: A Laboratory Manual 2nd ed. (Cold Spring Harbor Press, Cold Spring Harbor N.Y., 2013); Kohler and Milstein, (1976) Eur. J. Immunol. 6: 511; Queen et al. U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332: 323 (1988); Bird, Science 242:423-42 (1988); Tomlinson I. and Holliger P. (2000) Methods Enzymol, 326, 461-479; Holliger P. (2005) Nat. Biotechnol. September; 23(9):1126-36).
  • One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described. For purposes of the present invention, the following terms are defined below.
  • Non-limiting examples of “Biological sample” as used herein means any biological material from which nucleic acids and/or proteins can be obtained. As non-limiting examples, the term encompasses whole blood, peripheral blood, plasma, serum, saliva, mucus, urine, semen, lymph, fecal extract, cheek swab, cells or other bodily fluid or tissue, including but not limited to tissue obtained through surgical biopsy or surgical resection. Alternatively, a sample can be obtained through primary patient derived cell lines, or archived patient samples in the form of preserved samples, or fresh frozen samples.
  • “Treatment” and “treating” as used herein refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition, prevent the pathologic condition, pursue or obtain good overall survival, or lower the chances of the individual developing the condition even if the treatment is ultimately unsuccessful. Those in need of treatment include those already with the condition as well as those prone to have the condition or those in whom the condition is to be prevented.
  • “SNP” as used herein means single nucleotide polymorphism.
  • “Risk variant” as used herein refers to an allele, whose presence is associated with an increase in susceptibility to an inflammatory bowel disease, including but not limited to Crohn's Disease, Ulcerative Colitis and Medically Refractory-Ulcerative Colitis, relative to an individual who does not have the risk variant.
  • “IBD”, “CD”, “UC” and “MR-UC” as used herein refer to Inflammatory Bowel Disease, Crohn's Disease, Ulcerative Colitis and Medically Refractive Ulcerative Colitis, respectively.
  • As used herein, “IBD” includes “CD”, “UC” and/or “MR-UC”.
  • As used herein, “ANCA” means anti-neutrophil cytoplasmic antibodies.
  • As used herein, “OmpC” means outer membrane protein C.
  • As used herein, “eQTL” means expression quantitative trait loci.
  • As used herein, “mQTL” means methylation quantitative trait loci.
  • Non-limiting examples of RNASET2 SNPs are rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 and rs9366093.
  • Described herein are methods of diagnosing inflammatory bowel disease using RNASET2, TL1A and/or IFN-γ as a biomarker of disease severity in a patient population and selecting the patient population for anti-TL1A therapy. Further described are methods of treating these patient populations.
  • RNASET2 (ribonuclease T2) encodes an extracellular RNase and is the only member of the human Rh/T2/S family of acid ribonucleases (acid hydrolyses), which are only active in acidic pH. The optimal activity of RNASET2 is at pH 5 and it has a preferential cleavage of poly-A and poly-U. It contains two regions with catalytic function and demonstrates a cleavage preference near adenylic acid followed by guanylic acid. Three isoforms have been detected for RNASET2, the 27KD, 31KD and 36KD isoforms. The 27KD and 31KD isoforms are thought to result from proteolytic cleavage of the 36KD isoform. All three isoforms are glycosylated. Subcellular fractionation reveals that full length RNASET2 is located in the endoplasmic reticulum and the two smaller RNASET2 proteolytic products are located in the lysosome fraction. RNASET2 is highly conserved among the phyla, from viruses to humans, suggesting an important evolutionary function.
  • TL1A (TNFSF15) is a tumor necrosis factor family member expressed primarily on activated cells of the immune system, such as monocytes, macrophages, and dendritic cells, following stimulation by immune complexes or through interaction with enteric microorganisms. TL1A expression is enhanced in inflammatory bowel disease and higher TL1A levels are associated with disease severity. Genome-Wide Association Studies (GWAS) have identified TNFSF15 SNPs to be associated with IBD. Studies have shown that neutralizing TL1A antibodies attenuate colitis in murine colitis models, while constitutive TL1A expression depicts a worsened murine ileo-cecal inflammation and intestinal fibrostenosis.
  • IFN-γ plays a key role in the generation and perpetuation of mucosal inflammation in IBD. TL1A augments IL-12/IL-18-mediated IFN-γ secretion in PB T cells.
  • The inventors identify RNASET2, an IBD susceptibility gene, as a component of TL1A-mediated enhancement of IFN-γ production. Moreover, functional variants of RNASET2 are associated with a more ‘severe’ CD phenotype characterized by one or more disease flares and stricturing/penetrating disease behavior. Without being bound to any particular theory, the inventor believes that RNASET2 serves as a therapeutic biomarker associated with severe disease pathobiology and allows for the identification of a patient population most likely to benefit from therapy targeted to the TL1A-driven inflammatory pathway. The TNFSF15 disease-associated variants are correlated with increased and sustained expression of TL1A. TNFSF15 has been identified and confirmed in GWAS as an IBD-associated gene and is believed to play a role in modulating the location and severity of intestinal inflammation, as well as development of stricturing disease. Transgenic mice with constitutive expression of TL1A developed intestinal inflammation along with ileal and colonic fibrosis, which was reversed by anti-TL1A treatment. In UC, there is a strong association between development of a medically refractory disease and the TL1A locus. Although TL1A is an important pro-inflammatory cytokine associated with IBD pathogenesis, the molecular pathways underlying enhanced cytokine secretion and inflammation were poorly understood. Described herein, the inventors investigated the TL1A-dependent molecular triggers that induce cytokine expression, particularly IFN-γ, in T cells. This approach identified down-modulation of RNASET2 as a component of TL1A-mediated enhancement of IFN-γ production.
  • The inventors demonstrate a functional association of RNASET2 disease risk SNPs with decreased expression and hyper-methylation in T cells isolated from CD patients, as well as, an association with clinical parameters suggestive of complicated/resistant disease behavior and rapid recurrence of disease. The inventors show the regulatory potential for ETS TF in modulating RNASET2 expression and the involvement of homotypic T cell aggregation via ICAM1 as a component of RNASET2 mediated up-regulation of IFN-γ production. The data distinguish RNASET2 as a potential therapeutic biomarker and identify unique pathways for additional therapeutic modulation within a defined IBD population.
  • The inventors found that, in IBD patients, there was a significant inverse correlation between the expression of RNASET2 and TNFSF15. In addition, the inventors demonstrate a functional association between DNA hyper-methylation and decreased expression of RNASET2.
  • The inventors found that there was significant eQTL overlap with RNASET2 IBD risk alleles identified through GWAS in samples isolated from the peripheral T cells and small bowel surgical resections. Significant RNASET2 eQTL (rs429083) has been described in a recent report that measured autoimmune associated risk variants in whole thymic tissue samples. This SNP demonstrated the most significant eQTL in the data, as well. Moreover, the study provides clinically relevant evidence that decreased expression levels of RNASET2 were correlated in CD patients with clinical parameters suggestive of complicated and resistant disease. Notably, CD patients carrying the RNASET2 disease risk SNPs displayed an increase in development of stricturing/penetrating disease behavior. RNASET2 expression was significantly lower in T cells isolated from CD patients exhibiting one or more disease flares per year. Similarly, RNASET2 expression is decreased in small bowel mucosal samples, as well as, in peripheral samples from medically refractory CD patients (9 out of 11 which were non-responsive to anti-TNF therapy), requiring surgical intervention for disease management. Consistent with the finding, a recent study reported significant RNASET2 eQTL in whole blood from patients resistant to anti-TNF therapy. Moreover, RNASET2 disease-associated SNPs correlated with therapeutic failure of anti-TNF therapy, and intestinal resection of >40 cm clinical characteristic of overall disease severity. In patients with a history of multiple resections, RNASET2 disease risk SNP was associated with a faster time to reoperation. Likewise, RNASET2 disease associated SNPs were associated in patients with endoscopic recurrence characterized by a more severe (>2) Rutgeerts score, which without being bound to any particular theory, can be predictive for early clinical recurrence and need for reoperation.
  • The transcriptional regulatory regions and binding factors modulating RNASET2 expression are likewise poorly defined. The majority of disease associated variants identified by GWAS reside within regulatory non-coding regions corresponding to promoters or enhancer sequences. Without being bound to any particular theory, studies suggest that alteration in transcriptional regulation via disruption of transcription factor binding sites may play a role in the disease process. In the present study the inventors utilized TF motif analysis to prioritize and identify from the large number of variants demonstrating eQTL and mQTL a prospective regulatory SNP. The rs2149092 disease associated SNP alters the conserved ETS consensus binding sequence and likely disrupts binding of multiple overlapping TF binding sites including IRF4, SPI1 and ELF1. Moreover, there is a strong positive correlation between the levels of RNASET2 expression and ETS and JUN TF family members. Interestingly, IRF4, SPI1 and ELF1 have been implicated in T cell development and IRF4 and ELF1 have been associated by GWAS with IBD. Without being bound to any particular theory, these data support a functional role for rs2049092 as a modulator of TF-DNA interactions and set the stage for future studies to determine the mechanistic pathways by which TL1A attenuates expression of RNASET2 in disease.
  • In the present study the inventors describe a functional relationship between RNASET2 and the cell adhesion molecule, ICAM1. Enhanced IFN-γ secretion in response to TL1A was accompanied by a decrease in RNASET2 expression on the one hand and an increase in ICAM1 levels on the other. TL1A mediated IFN-γ secretion was inhibited by Ab blockade of the ICAM1-LFA1 interaction. Although ICAM1-LFA1 engagement is classically defined as occurring between endothelial and T cells, these interactions have more recently been shown to play a critical role in mediating homotypic cellular aggregation of activated T cells. Homotypic T-T aggregates have been shown to promote synaptic-based cytokine delivery of IFN-γ and IL2 from one T cell to another, resulting in IL-2 receptor ligation and subsequent STATS phosphorylation. The inventors demonstrate that enhanced cellular aggregation is a hallmark of IFN-γ producing cells and TL1A-stimulation increases the number and size of the cellular aggregates. Without being bound to any particular theory, these findings suggest that RNASET2 may act through the integrin signaling pathway to modulate downstream IFN-γ secretion.
  • In conclusion, the inventors identified a novel functional and biological relationship between two IBD susceptibility genes, TNFSF15 and RNASET2. The inventors provide evidence that decreased RNASET2 expression is functionally implicated in both the TL1A driven pro-inflammatory cytokine production by activated T cells and functionally associated with the RNASET2 IBD susceptibility variants. Likewise, the present study demonstrates an association between decreased RNASET2 expression and a more severe form of IBD inflammation, which without wishing to be bound by any particular theory, we believe underlie disease pathology triggered by TL1A mediated pathways. Decreased expression and altered epigenetic DNA methylation of RNASET2 characterize a subset of IBD patients with a more severe disease phenotype. The inventors demonstrate a functional association of RNASET2 disease risk SNPs with decreased expression and hyper-methylation in T cells isolated from CD patients, as well as, an association with clinical parameters suggestive of complicated/resistant disease behavior and rapid recurrence of disease. The inventors show the regulatory potential for ETS TF in modulating RNASET2 expression and the involvement of homotypic T cell aggregation via ICAM1 as a component of RNASET2 mediated upregulation of IFN-γ production. The data distinguish RNASET2 as a potential therapeutic biomarker and identify unique pathways for additional therapeutic modulation within a defined IBD population. Thus, RNASET2 expression serves as a novel disease biomarker of a more severe form of inflammation identifying a patient population not responsive to current treatment strategies, whom may benefit from alternate RNASET2 mediated therapeutic approaches.
  • As disclosed herein, the inventors have identified RNASET2 associated SNPs in an IBD patient cohort. The inventors have identified RNASET2 associated SNPs in a CD patient cohort. The inventors have identified RNASET2 associated SNPs in a UC patient cohort. The inventors have identified RNASET2 associated SNPs in a MR-UC patient cohort. The SNPs were associated with disease location, disease behavior and need for surgery. The inventors have further identified RNASET2 as a biomarker for disease severity and associated RNASET2 risk SNPs in an IBD patient cohort. RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a CD patient cohort. RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a UC patient cohort. RNASET2 has been identified as a biomarker for disease severity and associated RNASET2 risk SNPs in a MR-UC patient cohort. In addition, the inventors demonstrate a correlation between RNASET2, TL1A expression and IFN-γ secretion.
  • The present invention is based, at least in part, on these findings. The present invention addresses the need in the art for methods of diagnosing patients with IBD and identifying patients in need of treatment, using RNASET2, TL1A and/or IFN-γ. The invention further provides a process for patient identification and/or stratification.
  • Diagnosis
  • Various embodiments of the present invention provide for a method of diagnosing inflammatory bowel disease (IBD) in a subject, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; and diagnosing IBD in the subject based on the presence of one or more risk variants at the RNASET2 gene. In some embodiments, inflammatory bowel disease is Crohn's disease, ulcerative colitis or medically refractive ulcerative colitis. In various embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093. In various embodiments, the subject is diagnosed with IBD if 2, 3, 4, 5 or 6 RNASET2 risk variants as described herein are present. In various embodiments, the risk allele for rs2149085 is the T allele. In various other embodiments, the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085. In various embodiments, the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085, as described herein are present. In other embodiments, the presence of a greater number of risk variants in the sample indicates that the subject is in greater need of treatment. In some embodiments, the detection of RNASET2 risk variants is indicative of the need for treatment in the subject. In yet other embodiments, the subject is identified as needing anti-TL1A therapy. In various embodiments, the subject diagnosed with IBD demonstrates therapeutic failure of thiopurine and anti-TNF therapy. In various other embodiments, the subject diagnosed with IBD is determined to need surgical intervention. In some embodiments, the surgical intervention is intestinal resection.
  • TABLE 1
    RNASET2 risk variants
    SNP SEQ ID NO:
    rs1819333 1
    rs2149092 2
    rs9355610 3
    rs2149085 4
    rs1410295 5
    rs9366093 6
  • In other embodiments, the method of diagnosing inflammatory bowel disease (IBD) in a subject described herein comprises determining the expression level of RNASET2, TL1A and/or IFN-γ. In some embodiments, a subject with decreased RNASET2, and/or increased TL1A and/or IFN-γ levels is diagnosed with IBD. In various embodiments, inflammatory bowel disease is Crohn's disease. In various embodiments, inflammatory bowel disease is ulcerative colitis. In various embodiments, inflammatory bowel disease is medically refractive ulcerative colitis. In various embodiments, inflammatory bowel disease is a CD patient who required surgical intervention for disease management. In yet other embodiments, a subject with decreased RNASET2, and/or increased TL1A and/or IFN-γ levels is identified as a subject in need of a treatment that increases RNASET2, and/or decreases TL1A and/or IFN-γ. In other embodiments, the subject is identified as needing anti-TL1A therapy. In yet other embodiments, the subject is identified as needing a treatment that causes an increase in RNASET2. In certain other embodiments, the subject is identified as needing a treatment that causes a decrease in IFN-γ and/or TL1A.
  • In various embodiments, the detection of RNASET2 risk variants and/or RNASET2, TL1A and/or IFN-γ expression levels can be accomplished by analyzing nucleic acids of a biological sample from the subject. A variety of apparatuses and/or methods, including, without limitation, polymerase chain reaction based analysis, sequence analysis and electrophoretic analysis can be used to detect RNASET2 risk variants. The expression levels of RNASET2, TL1A and/or IFN-γ can be detected using a variety of apparatuses and/or methods, including, without limitation, quantitative PCR, northern blot and microarrays. As used herein, the term “nucleic acid” means a polynucleotide such as a single or double-stranded DNA or RNA molecule including, for example, genomic DNA, cDNA and mRNA. The term nucleic acid encompasses nucleic acid molecules of both natural and synthetic origin as well as molecules of linear, circular or branched configuration representing either the sense or antisense strand, or both, of a native nucleic acid molecule.
  • In various other embodiments, determining the expression level of RNASET2, TL1A and/or IFN-γ can be accomplished by analyzing the proteins of a biological sample from the subject. A variety of apparatuses and/or methods, including, without limitation, ELISA, immunohistochemistry, and western blot can be used to detect RNASET2, TL1A and/or IFN-γ expression levels.
  • Various embodiments of the present invention also provide for a method of diagnosing medically refractive ulcerative colitis (MR-UC), comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2; and diagnosing MR-UC in the subject based on the presence of one or more risk variants at the RNASET2 gene. In various embodiments, the one or more risk variants at the RNASET2 gene are rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093. In various embodiments, the risk allele for rs2149085 is the T allele. In various other embodiments, the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085. In various embodiments, the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present.
  • In various embodiments, the subject diagnosed with MR-UC demonstrates therapeutic failure of thiopurine and anti-TNF therapy. In various other embodiments, the subject diagnosed with MR-UC is determined to need surgical intervention. In some embodiments, the surgical intervention is intestinal resection. In various embodiments, the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • In various other embodiments, the methods further comprise determining the level of methylation of RNASET2, and diagnosing IBD in a subject who has an increase in RNASET2 methylation. In other embodiments, the level of RNASET2 methylation is determined to diagnose a subject with MR-UC. In some embodiments, the subject is identified as needing a treatment that causes a decrease in RNASET2 methylation. In other embodiments, the subject is identified as needing an anti-TL1A therapy.
  • Various embodiments of the present invention provide for the treatment of subjects diagnosed with MR-UC. MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine. In various embodiments, the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various embodiments, the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • In various embodiments, the subject is identified as needing a treatment that mimics, RNASET2, TL1A and/or IFN-γ. In other embodiments, the subject is identified as needing a treatment that modulates RNASET2, TL1A and/or IFN-γ. In some other embodiments, the subject is identified as needing a treatment that targets RNASET2, TL1A and/or IFN-γ. In yet other embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various embodiments, the disease is IBD. In various embodiments, the disease is CD. In various embodiments, the disease is UC. In various embodiments, the disease is MR-UC. In various embodiments, the subject diagnosed is a CD patients who required surgical intervention for disease management.
  • In various embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2. In other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2 in peripheral and mucosal tissues. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with DNA hypermethylation in patients requiring surgical intervention for disease management. In yet other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with therapeutic failure of thiopurine and/or anti-TNF therapy. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with ANCA sero-positivity. In various other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with increased overall length of intestinal resection.
  • Subject Identification and/or Stratification
  • Various embodiments of the present invention provide for a process of identifying a subject with inflammatory bowel disease for treatment, comprising: determining the expression level of RNASET2, TL1A and/or IFN-γ; and identifying the subject in need of treatment as a subject with decreased RNASET2, and/or increased TL1A and/or IFN-γ levels. In various embodiments, the inflammatory bowel disease is Crohn's disease. In various embodiments, the inflammatory bowel disease is ulcerative colitis. In various embodiments, the inflammatory bowel disease is medically refractive ulcerative colitis. In various other embodiments, the subject is identified as needing a treatment that causes an increase in RNASET2. In yet other embodiments, the subject is identified as needing a treatment that causes a decrease in TL1A and/or IFN-γ. In certain embodiments, the subject is identified as needing anti-TL1A therapy. In some embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides.
  • “Patient Risk Stratification” as used herein means the process of separating subjects into risk groups in need of treatment.
  • Various embodiments of the present invention provide for a process of patient risk stratification to identify a subject in need of treatment, relative to a healthy individual. In various embodiments, the subject is stratified based on the detection of RNASET2, TL1A and/or IFN-γ in a biological sample from the subject. In some embodiments, a decrease in RNASET2 is indicative of a patient having IBD, in need of treatment. In some embodiments, a decrease in RNASET2 is indicative of a patient having CD, in need of treatment. In some embodiments, the patient is a CD patient who requires surgical intervention for disease management. In some embodiments, a decrease in RNASET2 is indicative of a patient having UC, in need of treatment. In some embodiments, a decrease in RNASET2 is indicative of a patient having MR-UC in need of treatment. In various other embodiments, an increase in TL1A and/or IFN-γ is indicative of a patient having IBD, in need of treatment. In various other embodiments, an increase in TL1A and/or IFN-γ is indicative of a patient having CD, in need of treatment. In various other embodiments, an increase in TL1A and/or IFN-γ is indicative of a patient having UC, in need of treatment. In various other embodiments, an increase in TL1A and/or IFN-γ is indicative of a patient having MR-UC, in need of treatment. In certain other embodiments, a decrease in RNASET2, an increase in TL1A, and an increase in IFN-γ is indicative of a subject having IBD, in need of treatment. In certain other embodiments, a decrease in RNASET2, an increase in TL1A, and an increase in IFN-γ is indicative of a subject having CD, in need of treatment. In certain other embodiments, a decrease in RNASET2, an increase in TL1A, and an increase in IFN-γ is indicative of a subject having UC, in need of treatment. In certain other embodiments, a decrease in RNASET2, an increase in TL1A, and an increase in IFN-γ is indicative of a subject having MR-UC, in need of treatment. In various embodiments, detection of the genes provides a guide for the treatment of the subject. In certain embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various other embodiments, the process of patient risk stratification to identify a subject in need of treatment is relative to a healthy individual who has been previously treated. In various other embodiments, the process of patient risk stratification to identify a subject in need of treatment is relative to a medically responsive individual.
  • Various embodiments of the present invention provide for the treatment of subjects diagnosed with MR-UC. MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine. In various embodiments, the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various embodiments, the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • In other embodiments, the process for subject identification and/or stratification described herein comprises determining the presence of one or more risk variants. In certain embodiments, the one or more risk variants comprise rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093. In various embodiments, the process comprises identifying the subject with IBD in need of treatment if 2, 3, 4, 5 or 6 RNASET2 risk variants as described herein are present. In various embodiments, the risk allele for rs2149085 is the T allele. In various other embodiments, the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085. In various embodiments, the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present. In various other embodiments, the detection of the risk variants in the biological sample stratifies the subject into a group needing treatment. In other embodiments, the presence of a greater number of risk variants in the sample indicates that the subject is in greater need of treatment. In some embodiments, the detection of RNASET2 risk variants is indicative of the need for treatment in the subject. In some embodiments, the subject is identified as needing anti-TL1A therapy. In various embodiments, the subject identified as needing RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • In various embodiments, the detection of RNASET2 risk variants can be accomplished by analyzing nucleic acids of a biological sample from the subject, as discussed herein.
  • In other embodiments, the process for subject identification and/or stratification described herein further comprises assaying the sample to detect the level of RNASET2 methylation, relative to a healthy individual. In some embodiments, a subject with an increased level of RNASET2 methylation is identified as a subject in need of treatment. In some embodiments, the sample is assessed for the level of RNASET2 methylation and one or more RNASET2 risk variants. In certain embodiments, a subject who has an increase in RNASET2 methylation and the presence of one or more RNASET2 risk variants is identified as a subject in need of treatment. In other embodiments, the sample is assessed for the level of RNASET2 methylation and the expression levels of RNASET2, TL1A and/or IFN-γ. In certain embodiments, a subject who has an increase in RNASET2 methylation and a decrease in RNASET2, an increase in TL1A and/or IFN-γ is identified as a subject in need of treatment. In certain embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In some embodiments, the subject is identified as needing anti-TL1A therapy. In some embodiments, the subject is identified as needing a treatment that causes a decrease in RNASET2 methylation. In various other embodiments, the detection of an increase in RNASET2 methylation is indicative of a patient with severe CD requiring surgery. In a further embodiment, the subject is identified as needing a treatment that comprises colectomy and/or anti-TL1A therapy.
  • In various other embodiments, the process for subject identification and/or stratification described herein can further comprise assaying the sample to detect an increase or decrease of at least one microbial antigen (serological factor), relative to a healthy individual. In some embodiments, the microbial antigens (serological factors) assessed comprise ANCA, ASCA, OmpC, I2 and CBir. In some embodiments, the sample is assessed for one or more microbial antigens (serological factors) and one or more RNASET2 risk variants. In certain embodiments, a subject who has one or more risk serological factors and the presence of one or more RNASET2 risk variants is identified as a subject in need of treatment. In yet other embodiments, the sample is assessed for one or more risk serological factors and the expression levels of RNASET2, TL1A and/or IFN-γ. In certain embodiments, a subject who has one or more risk serological factors and a decrease in RNASET2, an increase in TL1A and/or IFN-γ is identified as a subject in need of treatment. In some embodiments, the subject is identified as needing a treatment that mimics, modulates and/or targets RNASET2, TL1A and/or IFNγ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In other embodiments, the treatment is anti-TL1A therapy. In various embodiments, the treatment is an RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • In various embodiments the subject identified with IBD demonstrates therapeutic failure of thiopurine and anti-TNF therapy. In various other embodiments, the subject identified with IBD is determined to need surgical intervention. In some other embodiments, the surgical intervention is intestinal resection. Various embodiments of the present invention also provide for a method of selecting surgery for a subject having Inflammatory Bowel Disease, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; diagnosing MR-UC in the subject based on the presence of one or more risk variants at the RNASET2 gene; and selecting surgery for the subject diagnosed with MR-UC. In some embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093. In various other embodiments, the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085. In various embodiments, the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present. In some embodiments, the method further comprises determining the level of methylation of RNASET2. In various embodiments, a subject with an increased level of RNASET2 methylation is identified as a subject in need of surgery. In other embodiments, the subject with an increase in RNASET2 methylation and the presence of one or more risk variants at the RNASET2 gene is identified as a subject in need of surgery.
  • In various embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2. In other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with decreased expression of RNASET2 in peripheral and mucosal tissues. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with DNA hypermethylation in patients requiring surgical intervention for disease management. In yet other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with therapeutic failure of thiopurine and/or anti-TNF therapy. In some other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with ANCA sero-positivity. In various other embodiments, the presence of one or more risk variants at the RNASET2 gene is associated with increased overall length of intestinal resection.
  • Various embodiments of the present invention provide for a method of selecting a therapy for a subject having Inflammatory Bowel Disease, comprising: obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; diagnosing medically refractive ulcerative colitis (MR-UC) in the subject based on the presence of one or more risk variants at the RNASET2 gene; and selecting surgery as the therapy and not selecting thiopurine or anti-TNF as the therapy for the subject diagnosed with MR-UC. In various embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610, rs2149085, rs1410295 or rs9366093. In various other embodiments, the RNASET2 risk variants are the RNASET2 risk variant rs429083 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the RNASET2 risk variants is one or more of the RNASET2 risk variants in Tables 2, 3, 4, 5, 6, 7, 8, 9, 10, and 13 and an RNASET2 risk variant selected from the group consisting of rs1819333, rs2149092, rs9355610, rs2149085, rs1410295, rs9366093, and combinations thereof. In various other embodiments, the one or more risk variants at the RNASET2 gene is rs1819333, rs2149092, rs9355610 or rs2149085. In various embodiments, the subject is diagnosed with IBD if 2, 3, or 4, RNASET2 risk variants rs1819333, rs2149092, rs9355610 or rs2149085 as described herein are present. In yet other embodiments, the method further comprises determining the level of methylation of RNASET2, wherein increased methylation is indicative of a subject requiring surgical intervention.
  • Various embodiments of the present invention provide for the treatment of subjects diagnosed with MR-UC. MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy, thiopurine therapy, corticosteroids and cyclosporine. In various embodiments, the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various embodiments, the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • Detection of Methylation
  • Various embodiments provide for a method of diagnosing a subject with inflammatory bowel disease (IBD). In some embodiments, the method comprises determining the level of methylation of RNASET2; and identifying the subject with IBD as a subject with increased RNASET2 methylation. In other embodiments, the method comprises identifying the subject with IBD as a subject who has increased RNASET2 methylation and who has the presence of one or more risk variants at the RNASET2 gene. In various other embodiments, the method comprises determining the expression level of RNASET2, TL1A and/or IFN-γ; and diagnosing the subject with IBD if the subject has a decrease in RNASET2, an increase in TL1A, an increase in IFN-γ and/or an increase in RNASET2 methylation.
  • Various embodiments provide for a process of identifying a subject with inflammatory bowel disease (IBD) in need of treatment. In some embodiments, the method comprises determining the level of methylation of RNASET2; and identifying the subject in need of treatment as a subject with increased RNASET2 methylation. In other embodiments, the expression level of RNASET2, TL1A and/or IFN-γ and the level of methylation of RNASET2 are determined, to identify a subject with inflammatory bowel disease in need of treatment. In various embodiments, the method comprises determining a subject in need of treatment as a subject who has a decrease in RNASET2, an increase in TL1A, an increase in IFN-γ, and/or an increase in RNASET2 methylation. In various other embodiments, the method comprises determining the presence or absence of one or more risk variants at the RNASET2 gene and identifying a subject with IBD in need of treatment as a subject who has an increase in RNASET2 methylation and the presence of one or more RNASET2 risk variants.
  • In various embodiments, an increase in methylation is indicative of a subject requiring surgical intervention. In yet other embodiments, an increase in RNASET2 methylation is indicative of requiring surgical intervention.
  • Various methods to detect levels of methylation include, but are not limited to the following assays, mass spectrometry, methylation-specific PCR (MSP), whole genome bisulfite sequencing, (BS-Seq), the HELP assay, ChIP-on-chip assays, restriction landmark genomic scanning, methylated DNA immunoprecipitation (MeDIP, MeDIP-chip, MeDIPseq), pyrosequencing of bisulfite treated DNA, molecular break light assay for DNA adenine methyltransferase activity, methyl sensitive southern blotting, separate native DNA into methylated and unmethylated fractions using MethylCpG Binding Proteins (MBPs) and/or Methyl Binding Domain (MBD), MethylationEPIC BeadChip, Illumina Infinium Methylation 450 BeadChip, High Resolution Melt Analysis (HRM or HRMA), and/or ancient DNA methylation reconstruction.
  • Various embodiments of the invention provide for the treatment of a subject diagnosed with inflammatory bowel disease (IBD) by the method comprising obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2 gene; and diagnosing IBD in the subject based on the presence of one or more risk variants at the RNASET2 gene. In various embodiments, inflammatory bowel disease is Crohn's disease, ulcerative colitis or medically refractive ulcerative colitis.
  • Various embodiments of the invention provide for the treatment of a subject diagnosed with medically refractive ulcerative colitis (MR-UC) by the method comprising, obtaining a sample from the subject; subjecting the sample to an assay adapted to determine the presence or absence of one or more risk variants at the RNASET2; and diagnosing MR-UC in the subject based on the presence of one or more risk variants at the RNASET2 gene.
  • Various embodiments of the present invention provide for the treatment of subjects diagnosed with MR-UC. MR-UC subjects are refractory to current conventional medical therapy used, such as anti-TNF therapy and thiopurine therapy. In various embodiments, the subjects diagnosed with MR-UC are treated with non-conventional treatments, such as, but not limited to treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ. In various embodiments, treatments that mimics, modulates and/or targets RNASET2, TL1A and/or IFN-γ can comprise antibodies and or silencing oligonucleotides. In various embodiments, the subject diagnosed with MR-UC is determined to need RNASET2 mediated therapy, such as but not limited to recombinant RNASET2 and anti-ICAM1. In various embodiments, the RNASET2 mediated therapy is an antibody or small molecule that targets genes that are upstream and/or downstream of RNASET2.
  • Biological Samples, Sample Preparation and Gene Expression Detection
  • In various embodiments, the steps involved in the current invention comprise obtaining a biological sample from a subject. The biological sample may be obtained either through surgical biopsy or surgical resection. Alternatively, a sample can be obtained through primary patient derived cell lines, or archived patient samples in the form of FFPE (Formalin fixed, paraffin embedded) samples, or fresh frozen samples. A sample may also comprise whole blood, peripheral blood, plasma, serum, saliva, cheek swab, or other bodily fluid or tissue. In various embodiments, the sample comprises tissue from the large and/or small intestine. In various other embodiments, the large intestine sample comprises the cecum, colon (the ascending colon, the transverse colon, the descending colon, and the sigmoid colon), rectum and/or the anal canal. In yet other embodiments, the small intestine sample comprises the duodenum, jejunum, and/or the ileum.
  • Nucleic acid or protein samples derived from the biological sample (i.e., tissue and/or cells) of a subject that can be used in the methods of the invention can be prepared by means well known in the art. For example, surgical procedures or needle biopsy aspiration can be used to collect the biological samples from a subject. In some embodiments, it is important to enrich and/or purify the abnormal tissue and/or cell samples from the normal tissue and/or cell samples. In other embodiments, the abnormal tissue and/or cell samples can then be microdissected to reduce the amount of normal tissue contamination prior to extraction of genomic nucleic acid or pre-RNA for use in the methods of the invention. Such enrichment and/or purification can be accomplished according to methods well-known in the art, such as needle microdissection, laser microdissection, fluorescence activated cell sorting, and immunological cell sorting.
  • Analysis of the nucleic acid and/or protein from an individual may be performed using any of various techniques. In various embodiments, assaying gene expression levels for RNASET2 comprises northern blot, reverse transcription PCR, real-time PCR, serial analysis of gene expression (SAGE), DNA microarray, tiling array, RNA-Seq, or a combination thereof. In various other embodiments, the gene expression levels for RNASET2, TL1A and/or IFN-γ are assayed. In other embodiments, the level of RNASET2 methylation is determined.
  • In various embodiments, methods and systems to detect protein expression include but are not limited to ELISA, immunohistochemistry, western blot, flow cytometry, fluorescence in situ hybridization (FISH), radioimmuno assays, and affinity purification.
  • The analysis of gene expression levels may involve amplification of an individual's nucleic acid by the polymerase chain reaction. Use of the polymerase chain reaction for the amplification of nucleic acids is well known in the art (see, for example, Mullis et al. (Eds.), The Polymerase Chain Reaction, Birkhauser, Boston, (1994)).
  • Methods of “quantitative” amplification are well known to those of skill in the art. For example, quantitative PCR involves simultaneously co-amplifying a known quantity of a control sequence using the same primers. This provides an internal standard that may be used to calibrate the PCR reaction. Detailed protocols for quantitative PCR are provided in Innis, et al. (1990) PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc. N.Y.). Measurement of DNA copy number at microsatellite loci using quantitative PCR analysis is described in Ginzonger, et al. (2000) Cancer Research 60:5405-5409. The known nucleic acid sequence for the genes is sufficient to enable one of skill in the art to routinely select primers to amplify any portion of the gene. Fluorogenic quantitative PCR may also be used in the methods of the invention. In fluorogenic quantitative PCR, quantitation is based on amount of fluorescence signals, e.g., TaqMan and sybr green.
  • Other suitable amplification methods include, but are not limited to, ligase chain reaction (LCR) (see Wu and Wallace (1989) Genomics 4: 560, Landegren, et al. (1988) Science 241:1077, and Barringer et al. (1990) Gene 89: 117), transcription amplification (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173), self-sustained sequence replication (Guatelli, et al. (1990) Proc. Nat. Acad. Sci. USA 87: 1874), dot PCR, and linker adapter PCR, etc.
  • A DNA sample suitable for hybridization can be obtained, e.g., by polymerase chain reaction (PCR) amplification of genomic DNA, fragments of genomic DNA, fragments of genomic DNA ligated to adaptor sequences or cloned sequences. Computer programs that are well known in the art can be used in the design of primers with the desired specificity and optimal amplification properties, such as Oligo version 5.0 (National Biosciences). PCR methods are well known in the art, and are described, for example, in Innis et al., eds., 1990, PCR Protocols: A Guide to Methods And Applications, Academic Press Inc., San Diego, Calif. It will be apparent to one skilled in the art that controlled robotic systems are useful for isolating and amplifying nucleic acids and can be used.
  • Hybridization
  • The nucleic acid samples derived from a subject used in the methods of the invention can be hybridized to arrays comprising probes (e.g., oligonucleotide probes) in order to identify RNASET2, TL1A and/or IFN-γ and in instances wherein a housekeeping gene expression is also to be assessed, comprising probes in order to identify selected housekeeping genes. In particular embodiments, the probes used in the methods of the invention comprise an array of probes that can be tiled on a DNA chip (e.g., SNP oligonucleotide probes). Hybridization and wash conditions used in the methods of the invention are chosen so that the nucleic acid samples to be analyzed by the invention specifically bind or specifically hybridize to the complementary oligonucleotide sequences of the array, preferably to a specific array site, wherein its complementary DNA is located. In some embodiments, the complementary DNA can be completely matched or mismatched to some degree as used, for example, in Affymetrix oligonucleotide arrays. The single-stranded synthetic oligodeoxyribonucleic acid DNA probes of an array may need to be denatured prior to contact with the nucleic acid samples from a subject, e.g., to remove hairpins or dimers which form due to self-complementary sequences.
  • Optimal hybridization conditions will depend on the length of the probes and type of nucleic acid samples from a subject. General parameters for specific (i.e., stringent) hybridization conditions for nucleic acids are described in Sambrook and Russel, Molecular Cloning: A Laboratory Manual 4th ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y. 2012); Ausubel et al., eds., 1989, Current Protocols in Molecules Biology, Vol. 1, Green Publishing Associates, Inc., John Wiley & Sons, Inc., New York, at pp. 2.10.1-2.10.16. Exemplary useful hybridization conditions are provided in, e.g., Tijessen, 1993, Hybridization with Nucleic Acid Probes, Elsevier Science Publishers B. V. and Kricka, 1992, Nonisotopic DNA Probe Techniques, Academic Press, San Diego, Calif.
  • Oligonucleotide Nucleic Acid Arrays
  • In some embodiments of the methods of the present invention, DNA arrays can be used to determine the expression levels of genes, by measuring the level of hybridization of the nucleic acid sequence to oligonucleotide probes that comprise complementary sequences. Various formats of DNA arrays that employ oligonucleotide “probes,” (i.e., nucleic acid molecules having defined sequences) are well known to those of skill in the art. Typically, a set of nucleic acid probes, each of which has a defined sequence, is immobilized on a solid support in such a manner that each different probe is immobilized to a predetermined region. In certain embodiments, the set of probes forms an array of positionally-addressable binding (e.g., hybridization) sites on a support. Each of such binding sites comprises a plurality of oligonucleotide molecules of a probe bound to the predetermined region on the support. More specifically, each probe of the array is preferably located at a known, predetermined position on the solid support such that the identity (i.e., the sequence) of each probe can be determined from its position on the array (i.e., on the support or surface). Microarrays can be made in a number of ways, of which several are described herein. However produced, microarrays share certain characteristics, they are reproducible, allowing multiple copies of a given array to be produced and easily compared with each other.
  • In some embodiments, the microarrays are made from materials that are stable under binding (e.g., nucleic acid hybridization) conditions. The microarrays are preferably small, e.g., between about 1 cm2 and 25 cm2, preferably about 1 to 3 cm2. However, both larger and smaller arrays are also contemplated and may be preferable, e.g., for simultaneously evaluating a very large number of different probes. Oligonucleotide probes can be synthesized directly on a support to form the array. The probes can be attached to a solid support or surface, which may be made, e.g., from glass, plastic (e.g., polypropylene, nylon), polyacrylamide, nitrocellulose, gel, or other porous or nonporous material. The set of immobilized probes or the array of immobilized probes is contacted with a sample containing labeled nucleic acid species so that nucleic acids having sequences complementary to an immobilized probe hybridize or bind to the probe. After separation of, e.g., by washing off, any unbound material, the bound, labeled sequences are detected and measured. The measurement is typically conducted with computer assistance. DNA array technologies have made it possible to determine the expression level of RNASET2, TL1A and/or IFN-γ, housekeeping genes and the methylation state of RNASET2.
  • In certain embodiments, high-density oligonucleotide arrays are used in the methods of the invention. These arrays containing thousands of oligonucleotides complementary to defined sequences, at defined locations on a surface can be synthesized in situ on the surface by, for example, photolithographic techniques (see, e.g., Fodor et al., 1991, Science 251:767-773; Pease et al., 1994, Proc. Natl. Acad. Sci. U.S.A. 91:5022-5026; Lockhart et al., 1996, Nature Biotechnology 14:1675; U.S. Pat. Nos. 5,578,832; 5,556,752; 5,510,270; 5,445,934; 5,744,305; and 6,040,138). Methods for generating arrays using inkjet technology for in situ oligonucleotide synthesis are also known in the art (see, e.g., Blanchard, International Patent Publication WO 98/41531, published Sep. 24, 1998; Blanchard et al., 1996, Biosensors And Bioelectronics 11:687-690; Blanchard, 1998, in Synthetic DNA Arrays in Genetic Engineering, Vol. 20, J. K. Setlow, Ed., Plenum Press, New York at pages 111-123). Another method for attaching the nucleic acids to a surface is by printing on glass plates, as is described generally by Schena et al. (1995, Science 270:467-470). Other methods for making microarrays, e.g., by masking (Maskos and Southern, 1992, Nucl. Acids. Res. 20:1679-1684), may also be used. When these methods are used, oligonucleotides (e.g., 15 to 60-mers) of known sequence are synthesized directly on a surface such as a derivatized glass slide. The array produced can be redundant, with several oligonucleotide molecules corresponding to each informative locus of interest (e.g., SNPs, RFLPs, STRs, etc.).
  • One exemplary means for generating the oligonucleotide probes of the DNA array is by synthesis of synthetic polynucleotides or oligonucleotides, e.g., using N-phosphonate or phosphoramidite chemistries (Froehler et al., 1986, Nucleic Acid Res. 14:5399-5407; McBride et al., 1983, Tetrahedron Lett. 24:246-248). Synthetic sequences are typically between about 15 and about 600 bases in length, more typically between about 20 and about 100 bases, most preferably between about 40 and about 70 bases in length. In some embodiments, synthetic nucleic acids include non-natural bases, such as, but by no means limited to, inosine. As noted above, nucleic acid analogues may be used as binding sites for hybridization. An example of a suitable nucleic acid analogue is peptide nucleic acid (see, e.g., Egholm et al., 1993, Nature 363:566-568; U.S. Pat. No. 5,539,083). In alternative embodiments, the hybridization sites (i.e., the probes) are made from plasmid or phage clones of regions of genomic DNA corresponding to SNPs or the complement thereof. The size of the oligonucleotide probes used in the methods of the invention can be at least 10, 20, 25, 30, 35, 40, 45, or 50 nucleotides in length. It is well known in the art that although hybridization is selective for complementary sequences, other sequences which are not perfectly complementary may also hybridize to a given probe at some level. Thus, multiple oligonucleotide probes with slight variations can be used, to optimize hybridization of samples. To further optimize hybridization, hybridization stringency condition, e.g., the hybridization temperature and the salt concentrations, may be altered by methods that are well known in the art.
  • In various embodiments, the high-density oligonucleotide arrays used in the methods of the invention comprise oligonucleotides corresponding to RNASET2, TL1A and/or IFN-γ and housekeeping genes. In other embodiments, the oligonucleotides correspond to methylated RNASET2. The oligonucleotide probes may comprise DNA or DNA “mimics” (e.g., derivatives and analogues) corresponding to a portion of each informative locus of interest (e.g., SNPs, RFLPs, STRs, etc.) in a subject's genome. The oligonucleotide probes can be modified at the base moiety, at the sugar moiety, or at the phosphate backbone. Exemplary DNA mimics include, e.g., phosphorothioates. For each SNP locus, a plurality of different oligonucleotides may be used that are complementary to the sequences of sample nucleic acids. For example, for a single informative locus of interest (e.g., SNPs, RFLPs, STRs, etc.) about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more different oligonucleotides can be used. Each of the oligonucleotides for a particular informative locus of interest may have a slight variation in perfect matches, mismatches, and flanking sequence around the SNP. In certain embodiments, the probes are generated such that the probes for a particular informative locus of interest comprise overlapping and/or successive overlapping sequences which span or are tiled across a genomic region containing the target site, where all the probes contain the target site. By way of example, overlapping probe sequences can be tiled at steps of a predetermined base interval, e. g. at steps of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 bases intervals. In certain embodiments, the assays can be performed using arrays suitable for use with molecular inversion probe protocols such as described by Wang et al. (2007) Genome Biol. 8, R246. For oligonucleotide probes targeted at nucleic acid species of closely resembled (i.e., homologous) sequences, “cross-hybridization” among similar probes can significantly contaminate and confuse the results of hybridization measurements. Cross-hybridization is a particularly significant concern in the detection of SNPs since the sequence to be detected (i.e., the particular SNP) must be distinguished from other sequences that differ by only a single nucleotide. Cross-hybridization can be minimized by regulating either the hybridization stringency condition and/or during post-hybridization washings. Highly stringent conditions allow detection of allelic variants of a nucleotide sequence, e.g., about 1 mismatch per 10-30 nucleotides. There is no single hybridization or washing condition which is optimal for all different nucleic acid sequences, these conditions can be identical to those suggested by the manufacturer or can be adjusted by one of skill in the art. In some embodiments, the probes used in the methods of the invention are immobilized (i.e., tiled) on a glass slide called a chip. For example, a DNA microarray can comprises a chip on which oligonucleotides (purified single-stranded DNA sequences in solution) have been robotically printed in an (approximately) rectangular array with each spot on the array corresponds to a single DNA sample which encodes an oligonucleotide. In summary the process comprises, flooding the DNA microarray chip with a labeled sample under conditions suitable for hybridization to occur between the slide sequences and the labeled sample, then the array is washed and dried, and the array is scanned with a laser microscope to detect hybridization. In certain embodiments there are at least 250, 500, 1,000, 2,000, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 11,000, 12,000, 13,000, 14,000, 15,000, 16,000, 17,000, 18,000, 19,000, 20,000, 21,000, 22,000, 23,000, 24,000, 25,000, 26,000, 27,000, 28,000, 29,000, 30,000, 31,000, 32,000, 33,000, 34,000, 35,000, 36,000, 37,000, 38,000, 39,000, 40,000, 41,000, 42,000, 43,000, 44,000, 45,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100,000 or more or any range in between, of RNASET2, TL1A and/or IFN-γ or housekeeping genes for which probes appear on the array (with match/mismatch probes for a single locus of interest or probes tiled across a single locus of interest counting as one locus of interest). The maximum number of RNASET2, TL1A and/or IFN-γ or housekeeping genes being probed per array is determined by the size of the genome and genetic diversity of the subjects species. DNA chips are well known in the art and can be purchased in pre-5 fabricated form with sequences specific to particular species. In other embodiments, SNPs and/or DNA copy number can be detected and quantitated using sequencing methods, such as “next-generation sequencing methods” as described further above.
  • Labeling
  • In some embodiments, the protein, polypeptide, nucleic acid, fragments thereof, or fragments thereof ligated to adaptor regions used in the methods of the invention are detectably labeled. For example, the detectable label can be a fluorescent label, e.g., by incorporation of nucleotide analogues. Other labels suitable for use in the present invention include, but are not limited to, biotin, iminobiotin, antigens, cofactors, dinitrophenol, lipoic acid, olefinic compounds, detectable polypeptides, electron rich molecules, enzymes capable of generating a detectable signal by action upon a substrate, and radioactive isotopes.
  • Radioactive isotopes include that can be used in conjunction with the methods of the invention, but are not limited to, 32P and 14C. Fluorescent molecules suitable for the present invention include, but are not limited to, fluorescein and its derivatives, rhodamine and its derivatives, texas red, 5′carboxy-fluorescein (“FAM”), 2′, 7′-dimethoxy-4′, 5′-dichloro-6-carboxy-fluorescein (“JOE”), N, N, N′, N′-tetramethyl-6-carboxy-rhodamine (“TAMRA”), 6-carboxy-X-rhodamine (“ROX”), HEX, TET, IRD40, and IRD41.
  • Fluorescent molecules which are suitable for use according to the invention further include: cyamine dyes, including but not limited to Cy2, Cy3, Cy3.5, CY5, Cy5.5, Cy7 and FLUORX; BODIPY dyes including but not limited to BODIPY-FL, BODIPY-TR, BODIPY-TMR, BODIPY-630/650, and BODIPY-650/670; and ALEXA dyes, including but not limited to ALEXA-488, ALEXA-532, ALEXA-546, ALEXA-568, and ALEXA-594; as well as other fluorescent dyes which will be known to those who are skilled in the art. Electron rich indicator molecules suitable for the present invention include, but are not limited to, ferritin, hemocyanin and colloidal gold.
  • Two-color fluorescence labeling and detection schemes may also be used (Shena et al., 1995, Science 270:467-470). Use of two or more labels can be useful in detecting variations due to minor differences in experimental conditions (e.g., hybridization conditions). In some embodiments of the invention, at least 5, 10, 20, or 100 dyes of different colors can be used for labeling. Such labeling would also permit analysis of multiple samples simultaneously which is encompassed by the invention.
  • The labeled nucleic acid samples, fragments thereof, or fragments thereof ligated to adaptor regions that can be used in the methods of the invention are contacted to a plurality of oligonucleotide probes under conditions that allow sample nucleic acids having sequences complementary to the probes to hybridize thereto. Depending on the type of label used, the hybridization signals can be detected using methods well known to those of skill in the art including, but not limited to, X-Ray film, phosphor imager, or CCD camera. When fluorescently labeled probes are used, the fluorescence emissions at each site of a transcript array can be, preferably, detected by scanning confocal laser microscopy. In one embodiment, a separate scan, using the appropriate excitation line, is carried out for each of the two fluorophores used. Alternatively, a laser can be used that allows simultaneous specimen illumination at wavelengths specific to the two fluorophores and emissions from the two fluorophores can be analyzed simultaneously (see Shalon et al. (1996) Genome Res. 6, 639-645). In a preferred embodiment, the arrays are scanned with a laser fluorescence scanner with a computer controlled X-Y stage and a microscope objective. Sequential excitation of the two fluorophores is achieved with a multi-line, mixed gas laser, and the emitted light is split by wavelength and detected with two photomultiplier tubes. Such fluorescence laser scanning devices are described, e.g., in Schena et al. (1996) Genome Res. 6, 639-645. Alternatively, a fiber-optic bundle can be used such as that described by Ferguson et al. (1996) Nat. Biotech. 14, 1681-1684. The resulting signals can then be analyzed to determine the expression of RNASET2, TL1A and/or IFN-γ and housekeeping genes, using computer software.
  • In other embodiments, where genomic DNA of a subject is fragmented using restriction endonucleases and amplified prior to analysis, the amplification can comprise cloning regions of genomic DNA of the subject. In such methods, amplification of the DNA regions is achieved through the cloning process. For example, expression vectors can be engineered to express large quantities of particular fragments of genomic DNA of the subject (Sambrook and Russel, Molecular Cloning: A Laboratory Manual 4th ed., Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y. 2012)).
  • In yet other embodiments, where the DNA of a subject is fragmented using restriction endonucleases and amplified prior to analysis, the amplification comprises expressing a nucleic acid encoding a gene, or a gene and flanking genomic regions of nucleic acids, from the subject. RNA (pre-messenger RNA) that comprises the entire transcript including introns is then isolated and used in the methods of the invention to analyze and provide a genetic signature of a cancer. In certain embodiments, no amplification is required. In such embodiments, the genomic DNA, or pre-RNA, of a subject may be fragmented using restriction endonucleases or other methods. The resulting fragments may be hybridized to SNP probes. Typically, greater quantities of DNA are needed to be isolated in comparison to the quantity of DNA or pre-mRNA needed where fragments are amplified. For example, where the nucleic acid of a subject is not amplified, a DNA sample of a subject for use in hybridization may be about 400 ng, 500 ng, 600 ng, 700 ng, 800 ng, 900 ng, or 1000 ng of DNA or greater. Alternatively, in other embodiments, methods are used that require very small amounts of nucleic acids for analysis, such as less than 400 ng, 300 ng, 200 ng, 100 ng, 90 ng, 85 ng, 80 ng, 75 ng, 70 ng, 65 ng, 60 ng, 55 ng, 50 ng, or less, such as is used for molecular inversion probe (MIP) assays. These techniques are particularly useful for analyzing clinical samples, such as paraffin embedded formalin-fixed material or small core needle biopsies, characterized as being readily available but generally having reduced DNA quality (e.g., small, fragmented DNA) and/or not providing large amounts of nucleic acids.
  • Once the expression levels have been determined, the resulting data can be analyzed using various algorithms, based on well-known methods used by those skilled in the art.
  • Kits
  • The present invention is also directed to a kit to diagnose a subject with IBD
  • and/or identifying a subject in need of treatment. The kit is useful for practicing the inventive method of diagnosing a subject and/or identifying a subject in need of treatment. The kit is an assemblage of materials or components, including at least one of the inventive compositions. Thus, in some embodiments the kit contains a composition including primers and probes for RNASET2, TL1A and/or IFN-γ, as described above.
  • The exact nature of the components configured in the inventive kit depends on its intended purpose. For example, some embodiments are configured for the purpose of assessing risk variants and/or gene expression levels. In some embodiments, the kit is configured to detect the gene expression levels of RNASET2 in a sample. In yet other embodiments, the kit is configured to detect the gene expression levels of RNASET2 and/or TL1A in a sample. In some other embodiments, the kit is configured to detect the gene expression levels of RNASET2, TL1A and/or IFN-γ in a sample. In various other embodiments, the kit is configured to detect RNASET2 risk variants in a sample. In yet other embodiments, the kit is configured to detect the level of RNASET2 methylation in a sample. In one embodiment, the kit is configured particularly for the purpose of assessing mammalian subjects. In another embodiment, the kit is configured particularly for the purpose of assessing human subjects. In further embodiments, the kit is configured for veterinary applications, assessing subjects such as, but not limited to, farm animals, domestic animals, and laboratory animals.
  • Instructions for use may be included in the kit. “Instructions for use” typically include a tangible expression describing the technique to be employed in using the components of the kit to effect a desired outcome, such as to diagnose a subject with IBD and/or identify a subject with IBD in need of treatment. Optionally, the kit also contains other useful components, such as, primers, diluents, buffers, pipetting or measuring tools or other useful paraphernalia as will be readily recognized by those of skill in the art.
  • The materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility. For example the components can be in dissolved, dehydrated, or lyophilized form; they can be provided at room, refrigerated or frozen temperatures. The components are typically contained in suitable packaging material(s). As employed herein, the phrase “packaging material” refers to one or more physical structures used to house the contents of the kit, such as inventive compositions and the like. The packaging material is constructed by well-known methods, preferably to provide a sterile, contaminant-free environment. The packaging materials employed in the kit are those customarily utilized in gene expression assays. As used herein, the term “package” refers to a suitable solid matrix or material such as glass, plastic, paper, foil, and the like, capable of holding the individual kit components. Thus, for example, a package can be a glass vial used to contain suitable quantities of an inventive composition containing primers and probes for RNASET2, TL1A, IFN-γ and/or RNASET2 methylation. The packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components.
  • EXAMPLES
  • The following examples are provided to better illustrate the claimed invention and are not to be interpreted as limiting the scope of the invention. To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention. One skilled in the art may develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention.
  • Example 1
  • Loss of RNASET2 in zebrafish results in accumulation of undigested rRNA within lysosomes. The major allele of RNASET2 (i) is a risk for IBD, CD (rs9355610), B1 and both ANCA levels and pos/neg (rs1410925) and (ii) protective for B3 and both ASCA IgA and IgG levels and pos/neg (rs1410925) (Table 2). The major allele rs9355610 is associated with lower levels of RNASET2 mRNA expression in CD small intestine and rectum, EBV transformed B cells and CD3+ PBL from IBD patients. The major allele is also associated with RNASET2 mRNA in CD sigmoid colon. Low levels of RNASET2 and increased levels of pANCA are associated with the major allele. Methylation at the RNASET2 locus is inversely correlated with RNASET2 mRNA expression.
  • TABLE 2
    Non-Jewish Qualitative and Quantitative Trait Association Data
    A1-
    iCHip CH BP minor Test NMISS OR STAT *P
    rs9355610 CD
    6 167383075 A ADD 3859 0.754 −4.041 5.33E−05 minor
    IBD
    6 167383075 A ADD 4172 0.8591 −2.607 9.12E−03 allele
    protective
    Al-
    Trait CH BP minor Test NMISS OR STAT P
    rs1410295 B1
    6 167345503 C ADD 538 0.7168 −2.588 9.64E−03 minor
    ANCA
    6 167345503 C ADD 875 0.7365 −2.626 8.65E−03 allele
    protective
    B3
    6 167345503 C ADD 537 1.408 2.337 1.95E−02 minor
    ASCA
    6 167345503 C ADD 820 1.379 2.926 3.44E−03 allele
    IgG risk
    ASCA
    6 167345503 C ADD 820 1.216 1.843 6.53E−02
    Panel
    BETA
    levels
    rs1410295 ANCA 6 167345503 C ADD 872 −4.608 −3.137 1.77E−03 minor
    allele
    protective
    ASCA
    6 167345503 C ADD 820 4.024 2.363 1.84E−02 minor
    IgA allele
    ASCA
    6 167345503 C ADD 820 5.745 2.821 4.90E−03 risk
    IgG
    All Non-Jewish A1 A2 Non-Jewish CD A1 A2
    CHR SNP Minor Major MAF CHR SNP Minor Major MAF
    6 rs1410295 C G 0.3541 6 rs1410295 C G 0.3484
    6 rs9355610 A G 0.3305 6 rs9355610 A G 0.2915
    *Bonferroni p-value 1.7E−04
  • TABLE 3
    RNASET2 Major allele is a risk for IBD and CD in a non-Jewish cohort
    Pheno Pheno Pheno
    SNP CH A1 A2 NMISS OR P Type OR P type OR P type
    rs9355610 6 A G 5913 0.8218 4.50E−05 IBD 0.756 1.22E−06 CD 0.94 0.3695 UC
    rs1819333 6 C A 5813 0.8523 3.91E−04 IBD 0.782 4.04E−06 CD 0.988 0.8575 UC
    *
    SNP CH A1 A2 MAF NCHROBS
    rs1819333 6 C A 0.4894 21278
    rs9355610 6 A G 0.3608 21278
    A1 = minor allele;
    A2 = major allele
  • TABLE 4
    Qualitative Trait Associations in non-Jewish CD. Major allele is risk for colonic
    disease in non-Jewish CD.
    CD- A1
    Phenotype CHR SNP BP A1 A2 NMISS OR STAT p Freq
    B1
    6 rs62436418 167265792 G A 592 0.6992 −2.853 4.33E−03 0.3787
    B2 6 rs62436418 167265792 G A 592 1.395 −2.648 8.09E−03
    Colon 6 rs62436418 167265792 G A 593 0.6275 −3.161 1.57E−03
    B2 6 rs9355610 167303065 A G 592 1.433 2.709 6.75E−03 0.3608
    Colon 6 rs9355610 167303065 A G 593 0.7299 −2.04 4.14E−02
    A1 = minor allele;
    A2 = major allele
  • TABLE 5
    RNASET2 SNPs Associated with CD and IBD. Major allele is risk for CD and IBD.
    Pheno
    type CHR SNP_rsid BP A1 A2 TEST NMISS OR STAT P LOCATION MAF
    CD
    6 rs2149085 167291100 G A ADD 4360 0.8414 −4.016 5.93E−05 INTERGENIC 0.4751
    CD 6 rs1819333 167293537 C A ADD 4361 0.8446 −3.93 8.51E−05 INTERGENIC 0.4753
    CD 6 rs3823208 167267836 G A ADD 4361 0.8507 −3.64 2.73E−04 INTRON 0.3446
    CD 6 rs62436418 167265762 G A ADD 4361 0.8569 −3.539 4.01E−04 INTERGENIC 0.3861
    CD 6 rs2769345 167286384 G A ADD 4361 0.8593 −3.53 4.15E−04 INTRON 0.4811
    CD 6 rs9459813 167287827 A T ADD 4361 0.8028 −3.104 1.91E−03 INTRON 0.1102
    CD 6 rs2236313 167280379 G A ADD 4361 0.8784 −3.016 2.56E−03 INTRON 0.4476
    CD 6 rs9355610 167303065 A G ADD 4361 0.9084 −2.163 3.05E−02 INTERGENIC 0.3502
    IBD 6 rs41269599 167267869 A G ADD 5110 0.844 −1.982 4.75E−02 INTRON 0.0635
    A1 = minor allele;
    A2 = major allele
  • TABLE 6
    RNASET2 SNPs Associated with Subclinical Phenotypes of CD
    PHENO
    TYPE CHR RSID BP A1 A2 TEST NMISS OR STAT P LOCATION MAF
    PDM
    6 rs3798303 167274358 G A ADD 2093 1.38 3.019 2.54E−03 INTRON 0.09853
    Iritis 6 rs41269599 167267869 A G ADD 1436 2.806 2.973 2.95E−03 INTRON 0.06876
    Iritis 6 rs181130555 167277010 D ADD 1434 2.744 2.932 3.37E−03 INTRON 0.07108
    Iritis 6 rs3777721 167272065 C G ADD 1436 2.761 2.92 3.50E−03 INTRON 0.07084
    Iritis 6 rs41269597 167267663 C G ADD 1436 2.711 2.868 4.13E−03 INTRON 0.07234
    Iritis 6 imm_6_167277028 167277028 A ADD 1435 2.609 2.776 5.51E−03 INTRON 0.07369
    Iritis 6 rs1079145 167280714 A G ADD 1436 2.424 2.717 6.59E−03 INTRON 0.0893
    Iritis 6 rs10946197 167268406 A C ADD 1436 0.3753 −2.717 6.60E−03 INTRON 0.256
    PDM 6 rs3777723 167273691 A G ADD 2093 1.334 2.648 8.09E−03 INTRON 0.09552
    A1 = minor allele;
    A2 = major allele;
    D = deletion
  • TABLE 7
    RNASET2 Associations with Subclinical Phenotypes in non-Jewish CD Patients
    PHENO
    TYPE CHR RSID BP A1 A2 TEST NMISS OR STAT P MAF
    Iritis
    6 rs1079145 167280714 A G ADD 680 3.646 2.99 2.79E−03 0.07357
    Iritis 6 rs3777721 167272065 C G ADD 680 3.844 2.97 2.98E−03 0.05932
    Iritis 6 rs181130555 167277010 D ADD 679 3.622 2.909 3.63E−03 0.0613
    Iritis 6 imm_6_167277028 167277028 A ADD 679 3.511 2.848 4.40E−03 0.06168
    Iritis 6 rs3778439 167278341 A G ADD 680 3.535 2.671 7.55E−03 0.05277
    Iritis 6 rs3734246 167278058 A G ADD 680 3.535 2.671 7.55E−03 0.05277
    PDM 6 rs2769339 167275844 G A ADD 977 1.553 2.929 3.41E−03 0.09707
    PDM 6 rs2757045 167270953 G A ADD 977 1.499 2.73 6.34E−03 0.09977
    PDM 6 rs3798303 167274358 G A ADD 977 1.491 2.669 7.62E−03 0.09669
    PDM 6 rs2757048 167271110 A G ADD 977 1.479 2.637 8.37E−03 0.099
    Uveitis 6 rs3777723 167273691 A G ADD 674 3.78 2.657 7.89E−03 0.09091
    A1 = minor alele;
    A2 = major allele;
    D = deletion
  • TABLE 8
    RNASET2 Association with Serologies in CD Patients
    BETA/
    Phenotype CHR SNP_rsid BP A1 A2 TEST NMISS OR STAT P LOCATION MAF
    CBIR
    6 rs425145 167321292 G A ADD 2432 4.175 2.167 3.03E−02 INTERGENIC 0.1121
    CBIR 6 rs415356 167326817 G A ADD 2432 4.16 2.164 3.06E−02 INTERGENIC 0.1121
    CBIR 6 rs435359 167317604 G A ADD 2432 4.16 2.164 3.06E−02 INTERGENIC 0.1123
    CBIR 6 rs375883 167304704 T A ADD 2430 4.119 2.14 3.24E−02 INTERGENIC 0.112
    CBIR 6 rs62438869 167320907 A G ADD 2432 −3.722 −2.101 3.57E−02 INTERGENIC 0.1281
    CBIR 6 rs9459813 167287827 A T ADD 2432 4.073 2.015 4.40E−02 INTRON 0.09872
    CBIR 6 rs443297 167304286 A G ADD 2432 3.978 1.972 4.88E−02 INTERGENIC 0.09947
    IgG.ASCA 6 rs1410295 167265493 C G ADD 2326 2.342 1.936 5.29E−02 INTERGENIC 0.3474
    A1 = minor allele;
    A2 = major allele
  • TABLE 9
    RNASET2 Associations with Serologies of non-Jewish CD Patients
    SEROLOGY CHR RSID BP A1 A2 TEST NMISS BETA STAT P MAF
    ANCA
    6 rs41269599 167267869 A G ADD 1143 −7.159 −2.709 6.85E−03 0.05586
    CBIR 6 rs9459813 167287827 A T ADD 1208 8.021 2.854 4.40E−03 0.1159
    CBIR 6 rs9459812 167287711 A C ADD 1208 7.699 2.735 6.33E−03 0.1156
    IgA.ASCA 6 rs3823208 167267836 G A ADD 1130 4.153 3.102 1.97E−03 0.3139
    IgG.ASCA 6 rs3823208 167267836 G A ADD 1130 4.99 3.115 1.89E−03 0.3139
    IgG.ASCA 6 imm_6_167277028 167277028 A ADD 1129 8.352 2.725 6.53E−03 0.06168
    A1 = minor allele.
    A2 = major allele
  • TABLE 10
    RNASET2 Associations with Expression in Small Bowel
    SNP_rsid gene beta p-value FDR
    rs72079749 RNASET2 0.151129328 1.66E−04 0.371075825
    rs66591848 RNASET2 0.154066135 2.27E−04 0.42289639
    rs1951459 RNASET2 0.161888591 2.39E−04 0.429556929
    rs4710149 RNASET2 0.161888591 2.39E−04 0.429556929
    rs933243 RNASET2 0.161888591 2.39E−04 0.429556929
    rs9355610 RNASET2 0.161888591 2.39E−04 0.429556929
    rs9356551 RNASET2 0.161888591 2.39E−04 0.429556929
    rs9366078 RNASET2 0.161888591 2.39E−04 0.429556929
    rs1819333 RNASET2 0.15479148 3.22E−04 0.461149469
    rs2013815 RNASET2 0.15479148 3.22E−04 0.461149469
    rs2149085 RNASET2 0.15479148 3.22E−04 0.461149469
    rs2769345 RNASET2 0.128393311 3.08E−03 0.777560209
    rs2236313 RNASET2 0.096154247 3.09E−02 0.908151161
    Major allele is risk for small bowel expression.
  • TABLE 11
    RNASET2 Associations for Expression in Large bowel
    snp_rsid gene beta t-stat p-value FDR
    CD
    Rectum rs683571 RNASET2 −0.57902 −3.65206 0.00115 0.460077
    Rectum rs2031846 RNASET2 −0.48099 −3.27469 0.002992 0.679028
    Rectum rs62436763 RNASET2 −0.50894 −3.24339 0.003235 0.692932
    Rectum rs41463945 RNASET2 −0.58473 −3.11594 0.004434 0.693762
    Sigmoid rs162289 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs162291 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs162293 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs162294 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs162295 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs162297 RNASET2 0.147287 2.955434 0.007311 0.619358
    Sigmoid rs2236312 RNASET2 0.162431 3.419281 0.002456 0.451404
    Sigmoid rs3756838 RNASET2 0.14602 3.147931 0.00467 0.576607
    Sigmoid rs3798307 RNASET2 0.162431 3.419281 0.002456 0.451404
    Sigmoid rs62436424 RNASET2 0.14602 3.147931 0.00467 0.576607
    Sigmoid rs7772112 RNASET2 0.14602 3.147931 0.00467 0.576607
    Sigmoid rs9366076 RNASET2 0.14602 3.147931 0.00467 0.576607
    UC
    Rectum rs57237533 RNASET2 0.572002 3.665898 0.001643 0.436982
    Rectum rs56213919 RNASET2 0.572002 3.665898 0.001643 0.436982
    Sigmoid rs10946198 RNASET2 −0.36397 −3.11588 0.008192 0.704927
    Sigmoid rs1819333 RNASET2 −0.36397 −3.11588 0.008192 0.704927
    Sigmoid rs2149085 RNASET2 −0.36397 −3.11588 0.008192 0.704927
    Sigmoid rs2769345 RNASET2 −0.36397 −3.11588 0.008192 0.704927

  • TABLE 13
    RNASET2 Exome CHIP
    Function Assoc
    dis SNP/rsID n.case n.ctrl CHR BP A1 A2 MAF NMISS OR P geneList GVS Locus
    CD exm- 5742 5725 6 1.7E+08 G A 0.4134 11467 0.8932 3.28E−05 RNASET2 intron IBD
    rs2236313
    IBD exm- 10523 5725 6 1.7E+08 G A 0.4145 16248 0.9219 0.00061 RNASET2 intron IBD
    rs2236313
    A1 = minor allele;
    A2 = major allele
  • Allele risk is defined by the Odds ratio (OR). When the A1 allele and an OR of <1 is depicted, then the major allele is the risk allele (A2 is risk). If the OR is >1, then the minor allele (A1) is risk allele. Knowing A1 and the Odds Ratio allows you to determine which allele is risk and which is protective.
  • Example 2
  • The molecular mechanisms of TL1A augmentation of inflammation via enhanced IFN-γ expression were defined using RNAseq. CD4+ T cells were analyzed in untreated conditions or treated with IL12 and IL18 or IL12 and IL18 and TLA1 at the 1 ug scale. On a 10 ng scale, CD4+ T cells were analyzed when treated with IL12 and IL18 and TLA1, either with or without IFN-γ. The RNAseq data prescreen removed all failed probe data, all genes with fewer than 3 samples with FPKM>5. Using this criteria, 8695 genes passed the prescreen and BRB Array Tools were used for class comparison using paired samples.
  • The inventors demonstrate that at the molecular level, TL1A treatment mediates enhanced expression of IFN-γ, in addition to mediating a decreased expression of RNASET2. The decreased expression of RNASET2 is detected in CD patients: 1) with chronically active disease, 2) with refractive disease requiring surgical intervention, 3) with patients naïve to anti-TNF therapy, 4) is associated with OmpC+, ANCA− serological factors, and 5) is associated with RNASET2 risk SNPs rs9355610, rs1819333, rs2149085.
  • Example 3
  • TNFSF15 and the protein it encodes TL1A, is associated with IBD and are key mediators of mucosal inflammation. In IBD patients, elevated TL1A levels correlate with disease severity and genotype. TL1A mediates marked enhancement of IFN-γ production. TL1A response biomarkers were identified by RNAseq and verified by qPCR in T cells isolated from IBD patients (20 Crohn's [CD], 20 ulcerative colitis [UC]) compared to normal (NL). An additional cohort of samples from NL and IBD patients was used to validate and measure expression/methylation quantitative trait loci (eQTL/mQTL) in the context of GWAS. RNAseq expression clustering differentiated TL1A treated versus non-treated cells. RNASET2, a gene encoding an extracellular T2 RNase, was down-regulated following TL1A treatment. Previous studies associated RNASET2 with susceptibility for CD. RNASET2 expression in CD patients was lower in “severe” vs. mild disease, i.e., multiple disease flare-ups (p<0.009), medically refractory (p<0.024). Disease risk allele for RNASET2 rs1819333 (p=0.015) and TNFSF15 allele associated with enhanced TL1A expression, rs6478108, rs6478109 and rs7848647 (p=0.01) were correlated with decreased RNASET2 expression. Moreover, siRNA silencing of RNASET2 enhanced TL1A mediated IFN-γ secretion. Without being bound to any particular theory, the inventors believe that down-regulation of RNASET2 is a hallmark of TL1A driven severe CD.
  • RNASET2 expression and DNA methylation were examined in a separate cohort of freshly isolated un-stimulated T cells from NL, CD or UC patients. Methylation at RNASET2 locus inversely correlated with mRNA expression. eQTL of RNASET2 alleles was associated with decreased expression. Significantly enhanced RNASET2 methylation was observed in CD patients with severe disease requiring surgical intervention (Table 14). No correlation was observed in NL or patients with mild disease. Likewise, increased RNASET2 methylation was associated with TNFSF15 risk alleles associated with enhanced TL1A expression (Table 14). Epigenetically, the RNASET2 eQTL/mQTL region overlaps with histone H3K4me3 and H3K27ac and DNase HS activation sites. This region co-localizes with transcription factor binding for NFκB, jun, ATF3 and CEBPD—all of which are up-regulated in response to TL1A treatment. The results identify RNASET2 as a TL1A response gene involved in regulation of IFN-γ production. In CD patients with severe disease there is hyper-methylation and decreased expression of RNASET2, which may be reflective of prior exposure in-vivo to TL1A. Thus, without being bound to any particular theory, the inventors believe that RNASET2 serves as a novel potential disease severity biomarker to identify a subset of CD patients most likely to benefit from anti-TL1A therapy.
  • TABLE 14
    Increased RNASET2 methylation associated with TNFSF15 risk allele.
    RNASET2 TNFSF15
    p value rs62436418 rs2236313 rs2769345 rs1819333 rs9355610 rs6478108 rs6478109 rs7848647
    eQTL 0.013 0.024 0.038 0.038 0.011 na na na
    mQTL 0.001 2.7 × 10−13 2.7 × 10−13 2.7 × 10−13 1.4 × 10−9 0.005 0.013 0.003
    (RNASET2)
  • The inventors further analyzed 3 cohorts of patients including 11 UC CD3+ PBT (medically refractive), 43 CD CD3+ PBT (23 were medically refractive and 20 were mild) and 17 normal CD3+ PBT. The samples were run on the Infinium 450 Methylation Array and 11 CD, 12 UC and 4 NL samples were run on the Infinium Expression Array. The inventors demonstrate that RNASET2 expression is decreased following TL1A treatment of CD4+ T cells and that silencing of RNASET2 enhances TL1A mediated IFN-γ secretion.
  • TABLE 15
    Clinical Features
    Expres- Methyla-
    RNASET2 sion tion
    Multiple disease flares
    Figure US20190300957A1-20191003-P00002
    Refractive disease requiring surgical intervention
    Figure US20190300957A1-20191003-P00002
    Figure US20190300957A1-20191003-P00003
    Patients naïve to anti-TNF therapy
    Figure US20190300957A1-20191003-P00002
    Figure US20190300957A1-20191003-P00003
    RNASET2 risk allele SNPs rs9355610, rs1819333,
    Figure US20190300957A1-20191003-P00004
    Figure US20190300957A1-20191003-P00005
    rs62436418, rs22236313, rs2769345
    RNASET2 risk allele SNPs
    Refractive disease
    Figure US20190300957A1-20191003-P00004
    Figure US20190300957A1-20191003-P00005
    Normal or Mild disease
    Figure US20190300957A1-20191003-P00006
  • Epigenetic studies demonstrated that RNASET2 eQTL/mQTL region overlaps with epigenetic activation sites for 1) histone H3K4me3 and H3K27ac, 2) DNase HS, 3) co-localizes with transcription factor binding for NFkB, jun, ATF3 and CEBPD, all of which are up-regulated in response to TL1A treatment. Furthermore, the enhancer element in primary T memory cells from peripheral blood, the DNAse HS site in CD4+ naïve T cells and the eQTL RPS6KA2 monocytes were linked to the rs1819333 allele.
  • Example 4
  • TL1A synergizes with IL-12/IL-18 resulting in a rapid (within 6-8 hours) and marked enhancement of IFN-γ expression. RNAseq analysis was used to identify the TL1A response genes regulating IFN-γ expression. Twenty genes were differentially expressed (at least 2-fold) in TL1A activated total CD4+ T cell population. This can be largely due to the fact that IFN-γ secreting T cells constitute only a very small subset (1-3%) of the total CD4+ T cell population (FIG. 15). CD4 T cells from healthy donors were treated with IL12/IL18 and TL1A for 8 hours and then sorted into IFN-γ-secreting and non-secreting subsets (FIG. 15) and whole-genome transcriptional analyses (GWAS) of mRNA was performed. Unsupervised hierarchical clustering of the entire 8075 expressed gene set clearly distinguished between the TL1A mediated IFN-γ-secreting and non-secreting subgroups (FIG. 16).
  • A class prediction analysis classifying the IFN-γ-secreting and non-secreting subgroups based on expression levels was performed. The best predictor transcript list consisted of 764 genes with at least two fold differential expression between the IFN-γ secreting subset (p-value<0.00005) (FIG. 17). Gene ontology analysis showed that the differentially expressed genes were enriched for those in pathways associated with proteasome, apoptosis, RNA expression and T cell receptor signaling, and were downstream targets of Infliximab (activation z score=−4, p value=2e-15). GWAS has identified multiple IBD risk variant SNPs. There was a significant increase in the proportion of transcripts located within 0.5 MB from an IBD risk SNPs (14% vs. 9%, p value=3.3e-6) compared to proportion of predictor genes in other regions. In fact, differentially expressed transcripts mapped to 34% of all IBD risk associated regions (FIG. 18). Without being bound to any particular theory, the data demonstrate a strong contribution for these genes not only in TL1A mediated modulation of IFN-γ expression but as contributing factors modulating IBD susceptibility and pathogenesis.
  • A volcano plot visualizing the significance and magnitude of differentially expressed predictor transcripts associated with IBD risk loci, allowed us to prioritize candidate genes (FIG. 20). Of these genes, TL1A mediated expression of IFN-γ was confirmed to be most significantly up-regulated and RNASET2 as most significantly down-regulated (FIG. 20). RNASET2, a member of the Rh/T2/S family of ribonuclease, was the only IBD risk associated gene displaying a greater than 5 fold TL1A mediated down-regulation in expression. RNASET2 has been identified by GWAS as a potential IBD risk gene. As the functional role of RNASET2 in IBD pathogenesis was unknown, the regulation of RNASET2 expression in IBD was examined. Without being bound to any particular theory, the inventors believe that since RNASET2 is a ‘class predictor’ gene, differential expression can be detected in total CD4+ T cells. Resting or IL12/IL18 treated CD4+ T cells from CD, UC patients or NL controls were isolated and RNASET2 levels were compared in the presence or absence of TL1A for 8 hours. As seen in FIG. 7, in contrast to what we had been observed in cells from NL donors, IBD patients did not display a TL1A mediated decrease in RNASET2 expression levels. Rather decreased expression levels of RNASET2 were associated in CD patients with “severe” compared to mild disease course. RNASET2 expression was significantly lower in cells isolated from CD patients exhibiting multiple disease flares per year (p<0.001) (FIGS. 8A and 8C) and likewise decreased RNASET2 expression was detected in CD patients who were medically refractive requiring surgical intervention for disease management (p<0.024) (FIG. 8B). A similar trend was seen in UC patients.
  • Gene expression quantitative trait (eQTL) was performed to characterize the functional correlation between RNASET2 gene variation and the gene transcript expression level. The IBD risk SNP tagging in Caucasians, rs1819333, and rs2149085 identified in the CD Korean population, are both located within −3.5 kb from the transcriptional start site, within the RNASET2 promoter region. An additional promoter SNP, rs9355610, has been shown to be associated with susceptibility with Grave's autoimmune thyroid disease. In peripheral cells isolated from CD patients the RNASET2 disease risk alleles rs1819333 (p=0.015) and rs2149085 (p=0.015), as well as, rs9355610 (p=0.04) display eQTL and were correlated with decreased RNASET2 expression (FIG. 9). Without being bound to any particular theory, the data indicates a pathway whereby down-regulation of RNASET2 alters IFN-γ expression. The functional role of RNASET2 in regulation of IFN-γ expression was confirmed using siRNA mediated silencing. CD4+ T cells were transfected with siRNA targeting RNASET2 mRNA or control siRNA and then treated with IL12/IL18 and TL1A. The expression of RNASET2 mRNA itself displayed a 60-70% inhibition by RNASET2 siRNA (FIG. 21A). In parallel, a significant enhancement (>1.5 fold) in IFN-γ expression was seen in cells transfected with RNASET2 siRNA compared to control scrambled siRNA (FIG. 21B).
  • Without being bound to any particular theory, the inventors believe that considering the pivotal role of IFNγ in pathogenesis of Crohn's disease these data collectively indicate that down-regulation of RNASET2 may serve as a biomarker of TL1A driven severe CD. RNASET2 expression was examined in a separate cohort of freshly isolated unstimulated T cells from NL, CD and UC patients. Since DNA methylation impacts upon gene expression and since most disease-risk genetic polymorphisms map outside of the transcribed exome in regions subject to epigenetic modification, the DNA methylation status of RNASET2 was examined as well. In unstimulated T cells from CD and UC patients an inverse correlation was observed between IFNγ expression levels and RNASET2 (FIG. 22). Moreover, as seen in FIG. 10, there was a significant negative correlation between expression and methylation which was inversely related to distance, mainly within 50 kb upstream and downstream from the transcriptional start site (TSS). Additionally, there was a significant overlap between IBD disease risk genetic variants and regions correlative for methylation and expression levels (FIG. 10). The strongest correlation (p=8.5×10−5) was observed at a CpG site (1.4 kb) within the first intron (FIG. 23).
  • The functional correlation between RNASET2 gene variation and the gene transcript expression level was confirmed in unstimulated peripheral T cells from IBD patients with refractory disease (FIG. 11), with decreased expression correlating with the RNASET2 risk allele variant SNP. Moreover, a similar eQTL was observed for mRNA extracted from tissue obtained from surgical resection of the small bowel using gene expression microarray (FIG. 12). The correlation between RNASET2 gene variation and methylation, mQTL was also examined and a significant mQTL was observed with an increase in methylation in IBD patients with refractory disease (FIGS. 13A-13D). In contrast, no mQTL was detected in cells isolated from patients with mild disease or NL subjects.
  • The role of these genetic variations affecting gene expression (eQTL) and DNA methylation (mQTL) was mapped across all informative SNPs spanning the RNASET2 locus (FIG. 14). In T cells isolated from IBD patients with medically refractory disease, there is strong overlapping eQTL and mQTL from 10 kb downstream of RNASET2 TSS to −170 Kb upstream, within the CCR6 locus. Likewise, there was a remarkable overlap in eQTL when comparing RNSASET2 expression from unstimulated peripheral T cells to small bowel surgical resection in patients with refractory disease. In contrast, in patients with mild disease or NL subjects no mQTL was detected (FIGS. 14A and 14B). This without being bound to any particular theory, the data suggests that down-modulation of RNASET2 is a component of TL1A mediated enhancement of IFN-γ expression. Additionally, epigenetic modulation of RNASET2 and reduced gene expression in IBD patients with a known IBD risk variant SNP is associated with a more severe course of disease.
  • Example 5 Methods
  • Peripheral T cells isolated from NL (normal) donors were cultured with or without TL1A for 8 hours. Gene expression profiling was performed by RNA sequencing (RNA-seq) and quantitative polymerase chain reaction (qPCR) using the subset of interferon gamma (IFN-γ)-producing cells purified by flow cytometry. Enzyme-linked immunosorbent assay (ELISA) and small interfering RNA (siRNA) inhibition and qPCR were used to measure the role of ribonuclease T2 (RNASET2) in TL1A mediated expression of IFN-γ. The role of RNASET2 in IBD was investigated using peripheral T cells isolated from IBD patients (20 CD and 20 UC) stimulated in a similar fashion. Findings were validated using additional samples of unstimulated T cells from NL and IBD patients or small bowel (SB) surgical resection and analyzed for expression quantitative trait loci (eQTL) and methylation quantitative trait loci (mQTL) based on genotyping and clinical data.
  • Screening for predicted motif disruption of transcription factor (TF) binding sites identified candidate regulatory SNPs. Proteomic analysis and measurement of cytokine secretion were used to determine effect of RNASET2-directed small interfering RNA (siRNA) on protein expression. Cell aggregation was measured by flow cytometry.
  • Study Subjects
  • Human subjects were recruited through the MIRIAD IBD Biobank at the F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute at Cedars-Sinai Medical Center. All control subjects were healthy individuals, free of medication, and with no known personal or family history of autoimmune disease or IBD. Informed consent (approved by the Institutional Review Board at Cedars-Sinai Medical Center) was obtained from all participating subjects. IBD patients were defined as “refractory” if surgical intervention was required for disease management following failure of medical therapy. IBD patients were defined as “mild” if they had no prior surgeries and no active disease at time of sample collection. CD patients exhibiting one or more disease flares per year were defined to have “severe disease” compared to patients with no disease flares per year. Clinical characteristics were prospectively collected from 564 CD patients who had undergone surgical resection.
  • Isolation of Lymphocyte Populations
  • Peripheral blood mononuclear cells (PBMC) were isolated from healthy volunteers by separation on Ficoll-Hypaque gradients. CD3+ T cells (PBT) were isolated using CD3-immunomagnetic beads (Miltenyi Biotech, Auburn, Calif.) and were at least 95% pure. CD4+ T cells were isolated using negative selection by depletion with magnetic beads (Stemcell Technologies, Vancouver, BC, Canada) and were at least 95% pure.
  • Infinium 450K Bead Chip Assay
  • DNA samples from CD3+ T cells were bisulfite converted using the Zymo EZ DNA Methylation kit (Zymo Research) with an input of 1 μg. The assay was carried out as per the Illumina Infinium Methylation instructions, using the Infinium HumanMethylation450 BeadChip Kit (Illumina Inc., San Diego, Calif.). Data were visualized and normalized using the GenomeStudio software. The methylation β values were recalculated as the ratio of (methylated probe signal)/(total signal).
  • IFN-γ Assay
  • IFN-γ was measured by an amplified ELISA. Greiner Bio-One (Longwood, Fla.) ELISA plates were coated overnight with 100 μl of 5 μg/ml monoclonal anti-IFN-γ (BD Biosciences, Woburn, Mass.). Samples and standards were added for 24 h followed by addition of 100 μl of 2.5 μg/ml polyclonal biotinylated rabbit anti-IFN-γ (BD Biosciences) for 2 h. This was followed by addition of 100 μl of 1/1000 diluted alkaline phosphatase-conjugated streptavidin (Jackson ImmunoResearch Laboratories, West Grove, Pa.) for 2 h. Substrate, 0.2 mM NADP (Sigma-Aldrich, St. Louis, Mo.) was added for 30 min followed by addition of amplifier (3% 2-propanol, 1 mM iodonitrotetrazolium violet, 75 μg/ml alcohol dehydrogenase, and 50 μg/ml diaphorase; Sigma-Aldrich) for 30 min. Plates were read at 490 nm using an E max plate reader (Molecular Devices, Sunnyvale, Calif.).
  • Gene Expression Assay for CD3+ T Cells
  • Expression analysis of CD3+ T cells was performed using the Illumina genome-wide expression BeadChip (HumanHT-12_V4_0_R2) (Illumina) or Nugen human FFPE RNA-seq library system. Illumina Gene expression data were processed using the BRB array tools and the lumi package in R. The data were log2-transformed and normalized using robust spline normalization. Libraries for RNA-Seq were prepared with Nugen human FFPE RNA-seq library system. The workflow consists of cDNA generation, fragmentation, end repair, adaptor ligation and PCR amplification. Different adaptors were used for multiplexing samples in one lane. Sequencing was performed on Illumina NextSeq 500 for a single read 75 run. Data quality check was done on Illumina SAV. Demultiplexing was performed with Illumina Bcl2fastq2 v 2.17 program. The reads were first mapped to the latest UCSC transcript set using Bowtie2 version 2.1.0 and the gene expression level was estimated using RSEM v1.2.15. FPKM was used to normalize the gene expression.
  • siRNA Inhibition and Quantitative Proteomic Analysis
  • Freshly isolated CD4+ T cells (15×106) were cultured overnight in RPMI 1640 medium containing 10% fetal calf serum, washed, resuspended in 250 μL fresh medium, and electroporated in the presence of 150 pmole of RNASET2 siRNA or control siRNA (600 V, for 9 pulses of 500 μsec, with 100 μsec between pulses) using 4 mm (gap width) cuvettes in a BTX Electro Square Porator ECM 830 (Genetronics, Inc., San Diego, Calif.). Sequences used in siRNA inhibition are depicted in Table 16.
  • TABLE 16
    siRNA sequences
    SEQ
    Sequence Name Sequence ID NO
    RNASET2 siRNA-sequence forward 5′-GCAAGAGAAAUUCACAAACUGCAGC-3′  7
    RNASET2 siRNA-sequence reverse 5′-GCUGCAGUUUGUGAAUUUCUCUUGCUU-3′  8
    Control siRNA-sequence forward 5′-CUUCCUCUCUUUCUCUCCCUUGUGA-3′  9
    Control siRNA-sequence reverse 5′-UCACAAGGGAGAGAAAGAGAGGAAGGA-3′ 10
  • Tandem mass tagging (TMT)-based quantitative proteomics analysis was conducted as described (Qu et al., Sci Rep 2016; 6:32007). For each sample, 50 μg proteins were digested in parallel into tryptic peptides using filter-aided sample preparation (FASP) (Wisniewski et al., Nat. Methods 2009; 6:359-62). Peptides derived from eight samples and a pooled internal standard were labeled with a set of TMT10plex reagents (Thermo Scientific), mixed, desalted, separated into 24 fractions by high-pH liquid chromatography, and concatenated into 8 fractions. Fractionated peptides were resolved on a 50 cm EASY-Spray analytical column, and analyzed by an LTQ Orbitrap Elite mass spectrometer (Thermo Scientific) in the data-dependent acquisition mode, using the higher-energy collisional dissociation (HCD) method for tandem mass spectrometry. Acquired raw data were searched against the human Uniprot database (released on Oct. 17, 2015, containing 20,982 sequences) with Proteome Discoverer (v2.1), using the SEQUEST algorithm. A stringent 1% false discovery rate was set to filter peptide and protein identifications. Peptides with >30% precursor ion interference were excluded from protein quantification.
  • Flow Cytometry and Analysis of Cellular Aggregation
  • IFN-γ-secreting CD4+ T cells was isolated by flow cytometry following activation of cells with recombinant human IL-12 (500 pg/ml, R&D Systems, Minneapolis, Minn.) and IL-18 (50 ng/ml, R&D Systems) and TL1A (100 ng/ml, Fitzgerald Industries International, Acton, Mass.) for 8 h. IFN-γ-secreting CD4+ T cells were detected using an IFN-γ secretion assay cell enrichment and detection kit (Miltenyi Biotec, San Diego, Calif.). Cells were sorted on a FACS Aria II (BD Biosciences, San Jose, Calif.).
  • Intracellular IFN-γ production and analysis of cellular aggregation was conducted essentially as described (Dezorella et al., Cytometry B Clin Cytom 2016:90:257-66) Briefly, cells were either rested or stimulated for 24 h with IL12/IL18 and TL1A and Berfeldin A (10 ug/ml) was added for the last 4 h. Cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 and 0.2% saponin and stained for intracellular IFN-γ (brilliant violet 421-IFN-γ, eBioscience) or isotype control. Samples were washed and stained for cellular aggregation (propidium iodide). Cells were acquired on a CyAnTM ADP Flowcytometer (Dako, Carpinteria, Calif., USA) and analyzed with FlowJosoftware (TreeStar Inc., Ashland, Oreg., USA). For LFA1 blocking analysis cells were pre-incubated overnight in conical microplates with monoclonal control mouse IgG1k (15 ug/ml) or anti-LFA1 (TS1/18) followed by stimulation with IL12, IL18 and TL1A for 24 h.
  • qPCR
  • Total RNA was isolated using the RNeasy kit (Qiagen, Inc., Valencia, Calif.) and gene expression was measured by real-time quantitative RT-PCR. Five hundred nanograms of total RNA were used in each RT-PCR reaction, with oligo (dT) (Integrated DNA Technologies) as primer, using the Omniscript kit and protocol (Qiagen). Real-time PCR was performed using a Mastercycler® ep realplex PCR detection system (Eppendorf, Hauppauge, N.Y.). PCR assays were run in duplicate. Primer sequences (Integrated DNA Technologies) spanned introns and are depicted in Table 17.
  • TABLE 17
    Primer sequences
    SEQ
    ID
    Sequence Name Sequence NO
    IFN-γ forward 5′-TTGGGTTCTCTTGGCTGTTACT-3′ 11
    IFN-γ reverse 5′-ATCCGCTACATCTGAATGACCTG-3′ 12
    RNASET2 5′-CTTCCTTGCAGGACTCACCAC-3′ 13
    forward
    RNASET2
    5′-GCTGATGTGAAGGTGCAAACTC-3′ 14
    reverse
    ACTB forward 5′-CGTGCTGCTGACCGAGG-3′ 15
    ACTB reverse 5′-AAGGTCTCAAACATGATCTGGGT-3′ 16
  • Genotyping
  • Genotype data was obtained for Caucasian subjects using Illumina HumanImmuno BeadChip array. Markers were excluded based on: test of Hardy-Weinberg Equilibrium with significance threshold of p≤10−3; if genotyping rate was <100% (for eQTL and mQTL associations) or <98% (for GWAS) and if minor allele frequency was <5%. Identity-by-descent was used to exclude related individuals (Pi-hat scores >0.25) using PLINK. ADMIXTURE was used to perform ethnicity analysis to get ethnicity proportion estimation for individuals. An individual with Caucasian proportion ≥0.75 was classified as Caucasian. Independent Caucasian samples were identified based on relatedness check (using cut-off pi-hat scores) and ethnicity analysis from admixture and all subsequent associations were performed using these samples. Principal components in genotype data for independent Caucasian samples were generated using TRACE. LDHeatmap R package was used to generate LD plot for the SNPs in RNASET2 locus using genotype data for 139 subjects. Details of the QC and genotyping in IIBDGC cohort can be found in previous reports (Jostins et al., Nature 2012; 491:119-24 and Liu et al., Nat Genet 2015; 47:979-86). In brief, 18,602 CD cases and 33,938 non-IBD controls genotyped using ImmunoChip were included in the analysis after samples with >5% missing data, samples of non-European ancestry from population stratification or with abnormal mean intensity values, and SNPs with >2% missing data or HWE p-value <10−10 in controls were removed. Of the CD cases from IIBDGC, 13,511 have disease behavior information collected based on Montreal classification as reported previously (Cleynen et al., Lancet 2016; 387:P156-67) (described as B1, non-stricturing, non-penetrating, B2, stricturing and B3, penetrating diseases).
  • Expression Data for Small Bowel Surgical Samples
  • Single channel microarray expression data extracted using Agilent feature extraction software were received from Genome Technology Access Center at Washington University, St. Louis. Raw expression data available in technical duplicates were normalized using LIMMA package implemented in R version 3.2.2. The expression data preprocessing included background correction of the expression data, followed by log 2-transformation and quantile-normalization.
  • EQTL and mQTL Mapping
  • eQTL and mQTL mapping was implemented in Matrix eQTL R package. For small bowel surgery samples, eQTL mapping was done using independent Caucasian samples (n=85). Associations between genotype and probe expression level (for eQTL) or methylation β values (for mQTL) were performed using a linear regression model with additive genotype effects. All associations were conducted with gender and first two principal components in genotype data as covariates along with genotype. Around 200 genetic variants within 200 KB of RNASET2 TSS were used to perform associations with RNASET2 gene expression or methylation levels.
  • Motif Analysis and Identification of Candidate Regulatory SNPs
  • All variants exhibiting eQTL and mQTL were analyzed for predicted disruption of TF binding motifs using the bioconductor motifbreakR package (Coetzee et al., Bioinformatics 2015; 31:3847-9). Only T cell specific TFs identified as being expressed using RNAseq data from CD patients, were carried forward. Candidate regulatory SNPs were then analyzed for potential functionality based on Roadmap Epigenomics Mapping Consortium (REMC) data (Roadmap Epigenomics C. et al, Nature 2015; 518:317-30). Potential active enhancer regions were determined based on overlap of the histone modification H3K4me1 with H3K27ac signals (Coetzee et al., Hum Mol Genet 2015; 24:3595-607). Potential functionality of TF regulation was determined based on REMC CHIP-seq binding signal and Regulome data.
  • Pathway Analysis
  • Pathway analysis was accomplished through the use of Qiagen's Ingenuity® Pathway Analysis (IPA®, Qiagen, Redwood City, www.qiagen.com/ingenuity) and The Database for Annotation, Visualization and Integrated Discovery (DAVID, http://david.abcc.ncifcrf.gov).
  • Statistical Analysis
  • Modeling, data analysis, and data mining were performed using the BRB array tools (brb.nci.nih.gov/BRB-ArrayTools) and R-program (version 2.2.2; www.r-project.org). Class prediction analysis used compound covariate predictor, diagonal linear discriminant analysis, k-nearest neighbor (using k=1 and 3), nearest centroid, and support vector machines, based upon a minimum p value of 0.001. Cluster analysis was performed using Cluster 3.0 and Java Treeview 1.1.6r4. Tests for statistical significance were determined using JMP Statistical Software (Cary, N.C.). Test for clinical association between of rs1819333 and rs9355610 SNPs and therapeutic failure, ANCA sero-positivity, resected bowel length and time to reoperation were calculated by parametric Student's T test and Pearson correlation; test of association and trend using Fisher's exact test and Kaplan-Meier Survival Curves. Association with endoscopic recurrence was calculated by Cochran-Armitage trend test.
  • Results
  • In this study, the inventors identified down-modulation of RNASET2, an IBD susceptibility gene, as a component of TL1A-mediated enhancement of cytokine production and as a novel potential biomarker of disease severity. Down-regulation of RNASET2 following siRNA silencing resulted in increased IFN-γ secretion, without being bound to any particular theory, supporting a role in regulating inflammatory response. A decrease in RNASET2 expression was also observed in peripheral T cells from CD patients with one or more yearly disease flares compared to patients with no yearly disease flares. Functionally, quantitative trait loci were associated with RNASET2 disease-risk variants for decreased expression (eQTL) in peripheral and mucosal tissues and DNA hypermethylation (mQTL) from CD patients with medically refractory, but not mild disease. Additionally, RNASET2 disease-risk variants were associated with an increase in development of stricturing/penetrating disease behavior. Furthermore, RNASET2 disease-risk variants were associated with a complicated/resistant CD phenotype defined in part by therapeutic drug failure, ANCA sero-positivity, increased length of intestinal resection, shorter time to reoperation and post-operative endoscopy with a high (>2) Rutgeerts score. Motif screening of RNASET2 disease-risk variants identified rs2149092 with predicted disruption of a consensus ETS-TF binding site located within a potential enhancer region, providing insight into RNASET2 cis-regulatory elements. RNASET2 correlated with expression of multiple ETS-transcription factors. Finally siRNA silencing of RNASET2 resulted in enhanced IFN-γ, increased ICAM1 and concomitant T cell aggregation while anti-LFA1 blocking of aggregation suppressed IFN-γ secretion.
  • Identification of Differential Gene Expression Associated with TL1A Mediated Enhancement of IFN-γ Production
  • To identify the underlying molecular pathways involved in TL1A-mediated enhancement of IFN-γ production, a key IBD proinflammatory cytokine, CD4+ T cells were treated with TL1A, sorted into IFN-γ-secreting and non-secreting subsets and analyzed by RNA-seq (FIG. 15 and FIG. 27). Unsupervised hierarchical clustering of the set of expressed genes clearly distinguished TL1A-mediated IFN-γ-secreting and non-secreting groups (FIG. 16). Seven hundred and sixty-four “predictor” genes with at least two-fold differential expression between the IFN-γ secreting/non-secreting subsets (p value <1×10−5) (FIG. 17) were identified. Gene ontology analysis indicated that differentially expressed genes were enriched in pathways associated with T cell receptor signaling, apoptosis, and RNA expression, and were downstream targets of infliximab, an anti-TNF biologic drug. Predictor genes were significantly enriched in regions flanking GWAS identified IBD susceptibility variants (0.25 MB upstream or downstream of the single nucleotide polymorphism (SNP) compared to other regions (14% vs. 9%, p value is 3.3×10−6, hypergeometric test). Without being bound to any particular theory, these data suggest that these genes contribute not only to TL1A-mediated modulation of IFN-γ expression, but also overlap with IBD risk-associated loci. Of the IBD-risk associated predictor genes, expression of IFN-γ was confirmed as the most significantly upregulated and RNASET2 as the most significantly down regulated gene (FIG. 20). RNASET2 was the only IBD risk associated gene with greater than 5-fold down regulation in the IFN-γ secreting CD4+ subset.
  • RNASET2 Regions Displaying Inverse Correlation of Expression and DNA Methylation Levels Overlap with Regions Flanking Disease-Risk Associated Variants
  • RNASET2 is the only human member of the Rh/T2/S family of ribonucleases and its expression is decreased in ovarian cancer, melanoma and non-Hodgkin lymphoma. Considering the key role for IFN-γ in pathogenesis of Crohn's disease/IBD, without being bound to any particular theory, these data collectively suggest that down regulation of RNASET2 identifies TL1A mediated ‘severe’ CD. RNASET2 expression was examined in freshly-isolated, unstimulated peripheral CD3+ T cells from a separate cohort of NL, CD and UC patients. Since DNA methylation is understood to be one of the mechanisms that impacts gene expression, particularly in disease-associated genetic variants that map outside transcribed exomes, we examined the DNA methylation status across the RNASET2 locus. RNA-seq analysis demonstrated there was an inverse correlation between TNFSF15 expression levels and RNASET2 in peripheral T cells from two independent cohorts comprised of a combined 138 CD patients (FIGS. 24A and 24D). These results were consistent even when each cohort was analyzed separately (FIGS. 24B and 24C). Moreover, there was a significant negative correlation between expression and methylation (FIG. 23), mainly within 50 kb upstream and downstream from the transcriptional start site (TSS) (FIG. 26). The strongest correlation of methylation and expression (p=8.5×10−5) was observed at a CpG site (1.4 kb) within the first intron of RNASET2 (FIG. 26). Additionally, CD disease genetic risk variants, including the IBD risk SNP tagging the RNASET2 locus in European ancestry populations, rs1819333, overlapped with regions correlative for methylation and expression levels (FIG. 10).
  • RNASET2 Disease-Risk Alleles are Associated with Decreased RNASET2 Expression and Increased DNA Methylation in CD Patients with Refractory Disease
  • Gene expression quantitative trait (eQTL) was performed to characterize the functional correlation between RNASET2 gene variation and the gene transcript expression level. The disease associated SNPs for IBD risk in Europeans, rs1819333, and Koreans, rs2149085, as well as the risk SNP associated with Graves' disease, rs9355610, are located −13 kb from the transcriptional start site of RNASET2. The functional correlation between RNASET2 IBD-risk genotypes and the gene transcript expression levels were established in unstimulated peripheral CD3+ T cells isolated from IBD patients with refractory disease. The data demonstrated significantly decreased RNASET2 expression in T cells from subjects carrying the RNASET2 risk alleles rs2149085, rs1819333, and rs9355610 (FIG. 11). These findings were confirmed with significant eQTL observed for mRNA extracted from uninflamed small bowel tissue obtained from CD subjects at surgical resection (FIG. 12). The correlation between RNASET2 gene variation and methylation, mQTL was also examined. A significant mQTL was observed with an increase in methylation in CD patients with refractory disease (FIGS. 13A and 13C). In contrast, no mQTL was detected in cells isolated from CD patients with mild disease or NL subjects (FIGS. 13B and 13D). Moreover, there was a significant increase of complicated disease behavior, stricturing/penetrating phenotype (Montreal classification B1 vs. B2 and B3), associated with CD patients carrying the RNASET2 disease risk SNPs (rs1819333/rs2149085 p=0.05, rs9355610 p=0.01).
  • Gene expression (eQTL) and DNA methylation (mQTL) was mapped across all informative SNPs spanning the RNASET2 locus (LD plot). In T cells isolated from patients with medically refractory disease, there is strong overlapping eQTL and mQTL from 10 kb downstream of RNASET2 TSS to −170 kb upstream, spanning fibroblast growth factor receptor 1 oncogene partner (FGFR1OP) to the first intron of chemokine (C—C motif) receptor 6 (CCR6). Likewise, there was overlap in eQTL when comparing RNASET2 expression from unstimulated peripheral T cells to small bowel surgical resection in CD patients with refractory disease. In contrast, few mQTL associations were detected in CD patients with mild disease or NL subjects (FIGS. 14A and 14B). No eQTL association was detected for FGFR1OP or CCR6. These data were further validated in a separate cohort of peripheral T cells isolated from CD patients with medically refractory disease. There was significant overlap between RNASET2 risk variants associated with CD and corresponding eQTL (FIG. 28) which, without being bound to any particular theory, suggest a functional role for RNASET2 in mediating disease.
  • Attenuated Expression of RNASET2 in CD
  • To establish a role for RNASET2 in IBD pathogenesis, regulation of RNASET2 expression in CD4+ T cells isolated from IBD patients was examined and compared to normal (NL) donors in the presence or absence of TL1A. As seen in FIG. 7, NL donors, but not IBD patients, exhibited a TL1A-mediated decrease in RNASET2 expression levels. Instead, decreased expression levels of RNASET2 were found in CD patients with more severe disease (exhibiting one or more disease flares per year) compared to patients with no yearly disease flares (FIGS. 8A and 8C).
  • RNASET2 Disease Risk Alleles are Associated with Complicated and Resistant Disease Behavior
  • To evaluate the association between RNASET2 and disease activity and severity the inventors utilized a cohort of 564 CD patients who had undergone surgical resection and were then followed prospectively. Clinical characteristics including indication for surgery were assessed for association with RNASET2 risk variants (rs1819333 and rs9355610). RNASET2 disease risk variant SNPs were associated with a complicated stricturing/penetrating phenotype (Montreal classification B1 vs. B2 and B3), (Table 18). At the time of surgery, patients with RNASET2 disease risk variant SNPs were associated with therapeutic failure of thiopurine or anti-TNF therapy, ANCA sero-positivity (a marker associated with lack of response to anti-TNF therapy), and an increased length of intestinal resection characteristic attributed to overall disease severity (Table 18 and FIGS. 38-39). No association was observed for therapeutic failure on steroids or sulfasalazine. Moreover, patients with RNASET2 disease risk variant SNPs who required multiple resections for disease management exhibited a shorter time to reoperation (FIG. 32).
  • TABLE 18
    Clinical disease parameters associated
    with RNASET2 risk variants.
    rs1819333 rs9355610
    Clinical parameter p OR p OR
    Disease Behavior
    B2 vs. B1a ns ns 0.041 1.07
    B3 vs. B1a ns ns 0.056 1.06
    B2, B3 vs. B1a 0.051 1.05 0.016 1.07
    Therapeutic failure of thiopurineb 0.009 1.68 0.019 1.75
    Therapeutic failure of anti-TNFb 0.039 1.46 0.042 1.56
    ANCA sero-positivityb 0.009 2.24 0.047 2.07
    Resected segment >30 cmb ns ns 0.004 2.13
    >40 cmb ns ns 0.031 1.96
    Endoscopic recurrence Rutgeert's
    score 3-4 vs 1-2 p z score p z score
    No prophylactic medsb 0.025 2.24 0.024 2.25
    aIIBDGC cohort CD (n = 3345)/case control (n = 6277)
    bCD patients (n = 584) who had undergone surgical resection and followed prospectively.
  • Likewise, RNASET2 risk SNPs were associated with a more severe disease recurrence. Post-operative endoscopies revealed an association of RNASET2 risk SNPs in patients classified with a high Rutgeerts score (>2) who were not receiving postoperative prophylaxis (Table 18). No association was observed for clinical recurrence. Decreased expression of RNASET2 was also associated with a penetrating disease phenotype (FIG. 31) and ASCA sero-positivity (FIG. 30). Without being bound by any particular theory, this data supports an association of RNASET2 disease risk SNPs with clinical parameters suggestive of complicated and resistant disease behavior.
  • RNASET2 Variant in LD with Disease-Tagging SNP Disrupts ETS Transcription Factor Binding Motif
  • The data presented above demonstrate significant overlap between more than a hundred CD RNASET2 risk variants, many in linkage disequilibrium, associated with eQTL and mQTL creating difficulty in determining functionality/causality. Since the majority of RNASET2 risk variants associated with CD are located in non-coding regions, it is likely that these SNPs alter expression through modulation of regulatory functions. Furthermore, without being bound to any particular theory, studies suggest that SNPs associated with disease often exist within active enhancer regions of cell types relevant to disease and can disrupt TF binding motifs. REMC data demonstrate that the RNASET2 locus is marked in primary T cells, compared to other tissues, by putative active enhancer histone modifications and active gene expression (FIG. 40). To gain insight into the molecular pathways regulating RNASET2 expression and prioritize the number of candidate functional SNPs, the inventors performed motif analysis to predict TF motif disruptions across all SNPs which were associated with eQTL/mQTL. Variants disrupting motifs of TFs expressed in T cells were selected and candidate variants in LD with the RNASET2 disease index SNP rs1819333 were focused on. The rs2049092 SNP disease risk variant, located −569 bp from the index SNP (LD R2=1), lies within the highly conserved TTCC motif, utilized by most ETS transcription factors, and is predicted to disrupt TF binding. Sequence analysis demonstrates an overlap of IRF4 and Spi1 binding sites adjacent to a JUN binding site (FIG. 36A). Regulome and REMC data confirm TF occupancy of ETS1, IRF4 and Spi1 binding in lymphoblastoid cell lines (FIG. 36B) which overlaps with histone modifications indicative of an active enhancer element. Moreover, there is a strong correlation between expression of RNASET2 with multiple members of ETS and JUN TF (FIGS. 36C and 41). No correlation was observed for IRF4 (FIG. 41). Without being bound to any particular theory, these data strengthen the relevance of RNASET2 expression in the immune compartment and support a functional role for ETS and JUN transcription factors in regulating transcription of RNASET2.
  • Silencing of RNASET2 Enhances IFN-γ Secretion Via Upregulation of ICAM1 Expression and Homotypic T Cell Aggregation
  • The functional role of RNASET2 in regulation of IFN-γ secretion was tested using siRNA silencing. CD4+ T cells transfected with siRNA targeting RNASET2 mRNA followed by stimulation with TL1A. Cells transfected with siRNA targeting RNASET2 displayed a 60-70% inhibition of RNASET2 expression (FIGS. 21A and 37A), and a parallel significant enhancement (>1.5 fold) in TL1A mediated IFN-γ secretion was seen compared to control siRNA (FIG. 21B and FIG. 37B). Without being bound to any particular theory, these data suggest that down regulation of RNASET2 modulates IFN-γ expression.
  • In order to define the signaling pathways involved in this process, proteomic analysis was carried out. Candidate targets were selected on the basis of exhibiting both modulation of expression following siRNA silencing of RNASET2 as well as TL1A-mediated differential expression when comparing IFNγ secreting and non-secreting T cells (data from RNAseq analysis). One of the proteins that was up-regulated in response to RNASET2 silencing and in the IFNγ secreting compared to non-secreting T cells, was ICAM1 (FIG. 42). ICAM1 was recently identified as an IBD susceptibility locus, with up-regulated gene expression associated with the disease-risk variant. ICAM1 is a transmembrane adhesion protein commonly expressed by vascular endothelium and leukocytes. Binding of ICAM to the LFA1 receptor on T cells facilitates and stabilizes cell-cell interactions. Studies have demonstrated increased ICAM1 expression on activated T cells and proposed a role for ICAM1-LFA1 binding in inducing homotypic T cell aggregation and subsequent T cells differentiation. To examine the effect of cell-cell contact on TL1A mediated IFN-γ secretion, cells were incubated in flat bottom and conical bottom microwells. A greater than 3 fold increase in IFN-γ production was consistently observed when cells were incubated in close cell-cell conical geometry (data not shown). Flow cytometry was then used to test the hypothesis that TL1A mediated enhancement of IFN-γ production is facilitated by homotypic T cell aggregation. Briefly, T cells were stimulated in the presence or absence of TL1A and then stained with an antibody for intracellular IFN-γ (FIGS. 37C and 37D, left panels) and for cellular aggregation using propidium iodide (PI) (FIGS. 37C and 37D, upper and lower right panels). The PI-labeled peaks correspond to number of cells per event allowing for identifying single cells versus cellular aggregates. The first peak in each histogram corresponds to single cell events (black brackets) and the successive peaks, to multicellular aggregates (gray brackets). Only a small percentage of the unstimulated T cells secreted IFN-γ, and these cells were almost equally distributed as single cell events and cellular aggregates (FIG. 37E, left panel). Following TL1A stimulation, there was a significant increase in both the percentage and size of cellular aggregates (upper right panels of FIG. 37C compared to FIG. 37D, as well as, the overall number of IFN-γ producing cells (6-fold) (FIGS. 37E and 37F) and a 30-fold increase in IFN-γ secretion (data not shown). In contrast, the majority of T cells that do not produce IFN-γ, are comprised of single cell events regardless of whether they were cultured with or without TL1A stimulation (FIG. 37E, right panel). Without being bound to any particular theory, these results suggest that cellular aggregation may contribute to both an increase in the number of cells producing IFN-γ and to overall amount of IFN-γ production, and TL1A stimulation may enhance this process. The functional role of TL1A in mediating cellular aggregation via ICAM1-LFA1 engagement was tested using an LFA-1 blocking antibody. As seen in FIG. 37G, there was an overall 43% reduction in IFN-γ secretion in response to blocking LFA-1 engagement, compared to IgG control antibody (p value=0.047). Without being bound to any particular theory, taken together these data indicate that TL1A-mediated downregulation of RNASET2 and concomitant enhancement of ICAM1 expression, promotes homotypic T cell aggregation and augmentation of IFN-γ production. It is noted that an increase in expression of ICAM1 was associated in CD with ASCA sero-positivity and pre-op therapeutic failure of anti-TNF and thiopurine (FIG. 43), clinical parameters associated with decreased RNASET2 and disease activity.
  • Example 6
  • Both RNASET2 and TNFSF15 have been identified among the 201 GWAS IBD susceptibility loci. TL1A, the protein encoded by TNFSF15, is a key mediator of mucosal inflammation. Elevated TL1A levels correlate with TNFSF15 genotype and disease severity. The inventors have identified that TL1A down regulates expression of RNASET2 in T cells. TNFSF15 and RNASET2 expression is inversely correlated in T cells from CD patients (p=5×10−16). The potential of RNASET2 as an IBD prognostic biomarker was examined.
  • The role for RNASET2 disease associated SNPs in IBD was analyzed by examining expression and methylation quantitative trait loci (eQTL/mQTL) in peripheral T cells from patients undergoing surgery (n=21) and small bowel surgical samples (n=85). CD patients (n=584) who had undergone surgical resection were followed prospectively. Clinical characteristics including indication for surgery were assessed for association with RNASET2 risk variants (rs1819333 and rs9355610).
  • RNASET2 disease associated SNPs were correlated with decreased RNASET2 expression (eQTL) in peripheral and mucosal tissues (p<0.001) and DNA hypermethylation (mQTL) (p<0.001) in patients requiring surgical intervention for disease management compared to those who were responsive to IBD therapeutics (n=16). RNASET2 disease associated SNPs were associated with therapeutic failure of thiopurine (p=0.02, OR=1.7) or anti-TNF therapy (p=0.04, OR=1.59), ANCA sero-positivity (a marker associated with lack of response to anti-TNF therapy) (p=0.02, OR=2.27), and an increase in overall length of intestinal resection (>30 cm p=0.004, OR=2.13/>40 cm p=0.03, OR=1.96). Patients with RNASET2 disease associated SNPs exhibited a shorter time to reoperation (p=0.04, z score=2.16). Post-operative endoscopies (n=369) with a high Rutgeerts score (>2) were associated with RNASET2 risk SNPs in patients not receiving post-op prophylaxis (p=0.02, z score=2.56) or those on anti-TNF therapy alone (p=0.03, z score=2.46), whereas no association was detected for patients on other IBD therapeutics.
  • This study identifies functional consequences of RNASET2 disease associated SNPs that are associated with clinically relevant disease behavior. RNASET2 risk SNPs were associated with clinical parameters suggestive of a complicated and resistant disease behavior. Moreover, response to therapeutics following surgery and recurrence of disease were associated with RNASET2 risk SNPs. Without being bound to any particular theory, these results taken together with our previous findings indicate that regulation of RNASET2 may underlie disease pathology triggered by TL1A and serve as a disease biomarker identifying subjects not responsive to current treatment strategies who may benefit from alternate therapeutic approaches.
  • Example 7
  • TABLE 19
    Clinical features associated with RNASET2 disease risk SNPs
    rs1819333 rs9355610
    CD phenotype at Time of Surgery p OR p OR
    therapeutic failure of thiopurine 0.009 1.68 0.019 1.75
    therapeutic failure of anti-TNF 0.039 1.46 0.042 1.59
    ANCA sero-positivity 0.009 2.24 0.047 2.07
    Length of resected segment >30 cm 0.004 2.13
    >40 cm 0.031 1.96
    Family history of disease 0.030 1.58 0.030 1.78
    CD patients (n = 584) who had undergone surgical resection and followed
    prospectively
    B2 vs B1 0.041 1.07
    B3 vs B1 0.056 1.06
    B2, B3 vs B1 0.051 1.05 0.016 1.07
    IIBDGC cohort CD (n = 7173)/case control (n = 6278)
  • TABLE 20
    Endoscopic Recurrence
    Endo- 3-year clinical
    scopic recurrence rate
    score Definition (%)
    0 No lesions 5
    1 aphtous lesions 5
    2 apthous lesions with normal mucosa between 15-20
    the lesions, or skip areas of larger lesions
    or lesions confined to ileocolonic anastomosis
    3 Diffuse apthous ileitis with diffusely 40
    inflamed mucosa
    4 Diffuse inflammation with already larger 90
    ulcers, nodules, and/or narrowing
    Adapted from Remedica Journals
    Post-op rs9355610
    Endoscopic recurrence Rutgeert's score 3-4 vs 1-2 p z score
    No prophylactic meds 0.040 2.25
    Anti-TNF alone 0.016 2.69
  • The rs2149092 (C-non-risk allele/T-risk allele) risk SNP abolishes IRF4/PU.1/ELF-1 binding site. IRF4 is an IBD susceptibility SNP that is lymphocyte specific and is essential for the differentiation of Th1, Th2, Th9, Th17 and T reg subsets. ELF-1 is a CD susceptibility SNP in the Japanese population. It is an ETS family transcription factor that is expressed in lymphoid cells, acts as both an enhancer and a repressor of expression and is involved in IL2 and IL23 signaling. PU.1 is also an ETS family transcription factor and is essential for early stages of T cell development. It down regulates γδ T Cells which are found in the mucosa and plays a role in innate immunity and when expressed in T H9 cells, these cells drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells.
  • RNASET2 expression is decreased following TL1A treatment in IFN-γ secreting CD4+ T cells and that silencing of RNASET2 enhances TL1A mediated IFN-γ secretion. Clinical correlates have also been identified for RNASET2 disease associated SNPs, which include, but are not limited to therapeutic failure of thiopurine therapy, therapeutic failure of anti-TNF therapy, ANCA Sero-positivity, B2/B3 versus B1 (structuring/penetrating vs non-penetrating/non-stricturing) disease, an increase in length of intestinal resection, decreased time to second surgery and endoscopic recurrence of disease with high Rutgeerts score.
  • Various embodiments of the invention are described above in the Detailed Description. While these descriptions directly describe the above embodiments, it is understood that those skilled in the art may conceive modifications and/or variations to the specific embodiments shown and described herein. Any such modifications or variations that fall within the purview of this description are intended to be included therein as well. Unless specifically noted, it is the intention of the inventors that the words and phrases in the specification and claims be given the ordinary and accustomed meanings to those of ordinary skill in the applicable art(s).
  • The foregoing description of various embodiments of the invention known to the applicant at this time of filing the application has been presented and is intended for the purposes of illustration and description. The present description is not intended to be exhaustive nor limit the invention to the precise form disclosed and many modifications and variations are possible in the light of the above teachings. The embodiments described serve to explain the principles of the invention and its practical application and to enable others skilled in the art to utilize the invention in various embodiments and with various modifications as are suited to the particular use contemplated. Therefore, it is intended that the invention not be limited to the particular embodiments disclosed for carrying out the invention.
  • While particular embodiments of the present invention have been shown and described, it will be obvious to those skilled in the art that, based upon the teachings herein, changes and modifications may be made without departing from this invention and its broader aspects and, therefore, the appended claims are to encompass within their scope all such changes and modifications as are within the true spirit and scope of this invention. It will be understood by those within the art that, in general, terms used herein are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.).

Claims (21)

1-34. (canceled)
35. A method of treating Inflammatory Bowel Disease (IBD) in a subject, the method comprising administering a therapeutically effective amount of a therapeutic agent to a subject, provided a single nucleotide polymorphism (SNP) at rs2149092 comprising a “T” at a nucleoposition 501 within SEQ ID NO: 2 is detected in a sample obtained from the subject.
36. The method of claim 35, provided a SNP at rs1819333 comprising an “A” allele at a nucleoposition 501 within SEQ ID NO: 1 is detected in the sample obtained from the subject.
37. The method of claim 36, wherein the IBD is severe IBD characterized by at least one of a therapeutic failure of an anti-Tumor Necrosis Factor alpha (TNFa) therapy, multiple flare-ups per year, stricturing disease, penetrating disease, and stricturing and penetrating disease.
38. The method of claim 36, provided a SNP at rs9355610 is detected in the sample obtained from the subject.
39. The method of claim 38, wherein the SNP at rs9355610 comprises a “G” allele at nucleoposition 501 within SEQ ID NO: 3.
40. The method of claim 36, provided a SNP at rs6478109 or a SNP at rs7848647, or a combination thereof, is detected in the sample obtained from the subject.
41. The method of claim 35, wherein the therapeutic agent comprises an anti-Tumor Necrosis Factor Ligand 1 (TL1A) therapy
42. The method of claim 35, wherein the therapeutic agent comprises a recombinant Ribonuclease T2 (RNASET2) polypeptide or protein.
43. The method of claim 35, wherein the IBD is Crohn's disease (CD), ulcerative colitis (UC), medically refractory UC (mrUC), or a combination thereof.
44. A method of diagnosing and treating inflammatory bowel disease in a subject, the method comprising:
a) subjecting a sample obtained from a subject to an assay adapted to determine a presence or an absence of a genotype comprising a single nucleotide polymorphism (SNP) rs2149092 comprising a “T” at a nucleoposition 501 within SEQ ID NO: 2;
b) detecting the presence or the absence of the genotype;
c) diagnosing inflammatory bowel disease (IBD) in the subject, provided the presence of the genotype is detected in step (b); and
d) treating the IBD in the subject, provided the subject is diagnosed with the IBD in (c)
45. The method of claim 44, wherein the genotype further comprises a SNP at rs1819333 comprising an “A” allele at a nucleoposition 501 within SEQ ID NO: 1.
46. The method of claim 44, wherein the IBD is severe IBD characterized by at least one of a therapeutic failure of an anti-Tumor Necrosis Factor alpha (TNFa) therapy, multiple flare-ups per year, stricturing disease, penetrating disease, and stricturing and penetrating disease.
47. The method of claim 44, provided the genotype further comprises a SNP at rs9355610 comprising a “G” allele at nucleoposition 501 within SEQ ID NO: 3.
48. The method of claim 44, wherein the assay comprises a genotyping assay selected from the group consisting of quantitative polymerase chain reaction (qPCR), a single nucleotide polymorphism (SNP) array, and sequencing.
49. The method of claim 44, provided the genotype further comprises at least one of a SNP at rs6478109 and a SNP at rs7848647.
50. The method of claim 44, wherein treating the IBD in (d) comprises administering to the subject a therapeutically effective amount of a therapeutic agent.
51. The method of claim 50, wherein the therapeutic agent comprises an anti-Tumor Necrosis Factor Ligand 1 (TL1A) therapy.
52. The method of claim 50, wherein the therapeutic agent comprises a recombinant Ribonuclease T2 (RNASET2) polypeptide or protein.
53. The method of claim 44, wherein the IBD is Crohn's disease, ulcerative colitis (UC), medically refractory UC, or a combination thereof.
54. A kit comprising:
a. a first nucleic acid probe comprising a first label, the first nucleic acid probe capable of hybridizing to at least a portion of a nucleic acid sequence provided in SEQ ID NO: 1 between nucleobases 300-700, including an “A” allele at nucleoposition 501; and
b. a second nucleic acid probe comprising a second label, the second nucleic acid probe capable of hybridizing to at least a portion of a nucleic acid sequence provided in SEQ ID NO: 2 between nucleobases 300-700, including a “T” allele at nucleoposition 501.
US16/084,858 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through RNASET2 Active 2037-09-19 US11186872B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/084,858 US11186872B2 (en) 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through RNASET2

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662309817P 2016-03-17 2016-03-17
US201762457048P 2017-02-09 2017-02-09
US16/084,858 US11186872B2 (en) 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through RNASET2
PCT/US2017/023082 WO2017161342A1 (en) 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through rnaset2

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/023082 A-371-Of-International WO2017161342A1 (en) 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through rnaset2

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/513,693 Continuation US20220162703A1 (en) 2016-03-17 2021-10-28 Methods of diagnosing inflammatory bowel disease through rnaset2

Publications (2)

Publication Number Publication Date
US20190300957A1 true US20190300957A1 (en) 2019-10-03
US11186872B2 US11186872B2 (en) 2021-11-30

Family

ID=59851075

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/084,858 Active 2037-09-19 US11186872B2 (en) 2016-03-17 2017-03-17 Methods of diagnosing inflammatory bowel disease through RNASET2
US17/513,693 Pending US20220162703A1 (en) 2016-03-17 2021-10-28 Methods of diagnosing inflammatory bowel disease through rnaset2

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/513,693 Pending US20220162703A1 (en) 2016-03-17 2021-10-28 Methods of diagnosing inflammatory bowel disease through rnaset2

Country Status (6)

Country Link
US (2) US11186872B2 (en)
EP (1) EP3430172A4 (en)
JP (2) JP7082945B2 (en)
KR (3) KR20180127416A (en)
CN (1) CN109462996A (en)
WO (1) WO2017161342A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021247770A1 (en) * 2020-06-03 2021-12-09 Cedars-Sinai Medical Center Methods and systems for measuring post-operative disease recurrence
US11236393B2 (en) 2008-11-26 2022-02-01 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-TNFα therapy in inflammatory bowel disease
US11312768B2 (en) 2013-07-19 2022-04-26 Cedars-Sinai Medical Center Signature of TL1A (TNFSF15) signaling pathway
WO2022140283A1 (en) * 2020-12-21 2022-06-30 Cedars-Sinai Medical Center Tl1a therapy compositions and methods of treatment therewith

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20210157418A (en) 2013-03-27 2021-12-28 세다르스-신나이 메디칼 센터 Mitigation and reversal of fibrosis and inflammation by inhibition of tl1a function and related signaling pathways
EP3430172A4 (en) 2016-03-17 2019-08-21 Cedars-Sinai Medical Center Methods of diagnosing inflammatory bowel disease through rnaset2
TW202012635A (en) * 2018-04-24 2020-04-01 美國錫安山醫學中心 Methods and systems for characterizing severe crohn's disease
CN109266749A (en) * 2018-11-23 2019-01-25 皖南医学院 A kind of primer and its detection method detecting RNASET2 gene promoter methylation
CA3121162A1 (en) * 2018-11-29 2020-06-04 Cedars-Sinai Medical Center Rnaset2 compositions and methods of treatment therewith
EP3920953A4 (en) * 2019-02-08 2022-12-14 Cedars-Sinai Medical Center Methods, systems, and kits for treating inflammatory disease targeting il18r1
TW202102538A (en) * 2019-03-26 2021-01-16 美商阿斯特捷利康合作創業有限責任公司 Patient populations amenable to il23-antagonist therapy
EP3748017A1 (en) * 2019-06-07 2020-12-09 Academisch Medisch Centrum Methylation markers for predicting sensitivity to treatment with antibody based therapy
CN113436741B (en) * 2021-07-16 2023-02-28 四川大学华西医院 Lung cancer recurrence prediction method based on tissue specific enhancer region DNA methylation

Family Cites Families (346)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3654090A (en) 1968-09-24 1972-04-04 Organon Method for the determination of antigens and antibodies
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US4016043A (en) 1975-09-04 1977-04-05 Akzona Incorporated Enzymatic immunological method for the determination of antigens and antibodies
US4265823A (en) 1979-01-04 1981-05-05 Robert E. Kosinski Aurothiosteroids
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
JPS60174629A (en) 1984-02-20 1985-09-07 Mitsubishi Monsanto Chem Co Manufacture of biaxially oriented polyamide film
US4699880A (en) 1984-09-25 1987-10-13 Immunomedics, Inc. Method of producing monoclonal anti-idiotype antibody
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4704692A (en) 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US5284931A (en) 1987-05-04 1994-02-08 Dana Farber Cancer Institute Intercellular adhesion molecules, and their binding ligands
US4935234A (en) 1987-06-11 1990-06-19 Dana-Farber Cancer Institute Method of reducing tissue damage at an inflammatory site using a monoclonal antibody
US5219997A (en) 1987-07-06 1993-06-15 Dana-Farber Cancer Institute Monoclonal antibody which inhibits the adhesion functions of the β integrin, CR3
US5114842A (en) 1987-07-08 1992-05-19 The Scripps Research Institute Peptides and antibodies that inhibit platelet adhesion
US4925572A (en) 1987-10-20 1990-05-15 Pall Corporation Device and method for depletion of the leukocyte content of blood and blood components
US4880548A (en) 1988-02-17 1989-11-14 Pall Corporation Device and method for separating leucocytes from platelet concentrate
US5147637A (en) 1988-06-07 1992-09-15 The Rockefeller University Method of inhibiting the influx of leukocytes into organs during sepsis or other trauma
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5235049A (en) 1989-01-24 1993-08-10 Molecular Therapeutics, Inc. Nucleic acid sequences encoding a soluble molecule (SICAM-1) related to but distinct from ICAM-1
US5002873A (en) 1989-03-17 1991-03-26 Fred Hutchinson Cancer Research Center DNA sequence encoding a lymphocyte adhesion receptor for high endothelium
US5091302A (en) 1989-04-27 1992-02-25 The Blood Center Of Southeastern Wisconsin, Inc. Polymorphism of human platelet membrane glycoprotein iiia and diagnostic and therapeutic applications thereof
US5272263A (en) 1989-04-28 1993-12-21 Biogen, Inc. DNA sequences encoding vascular cell adhesion molecules (VCAMS)
US5137806A (en) 1989-12-11 1992-08-11 Board Of Regents, The University Of Texas System Methods and compositions for the detection of sequences in selected DNA molecules
US5264554A (en) 1990-01-19 1993-11-23 The Blood Center Of Southeastern Wisconsin, Inc. Platelet cell adhesion molecule and variants thereof
GB9009548D0 (en) 1990-04-27 1990-06-20 Celltech Ltd Chimeric antibody and method
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5210015A (en) 1990-08-06 1993-05-11 Hoffman-La Roche Inc. Homogeneous assay system using the nuclease activity of a nucleic acid polymerase
WO1992002819A2 (en) 1990-08-10 1992-02-20 The Regents Of The University Of California Assay for ulcerative colitis and primary sclerosing cholangitis
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5085318A (en) 1990-11-19 1992-02-04 Leverick Kathy L Secured disc folder
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
ZA924276B (en) 1991-06-11 1993-03-31 Blood Res Center Intercellular adhesion molecule-3 and its binding ligands
EP1400536A1 (en) 1991-06-14 2004-03-24 Genentech Inc. Method for making humanized antibodies
US5227369A (en) 1991-07-11 1993-07-13 The Regents Of The University Of California Compositions and methods for inhibiting leukocyte adhesion to cns myelin
US5234810A (en) 1991-09-20 1993-08-10 The United States Of America As Represented By The Secretary Of Agriculture Diagnostic assays for genetic mutations associated with bovine leukocyte adhesion deficiency
WO1993007485A1 (en) 1991-10-10 1993-04-15 The Regents Of The University Of California Rare immunoglobulin framework regions associated with human disease
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES.
GB9126918D0 (en) 1991-12-19 1992-02-19 Univ London Members of specific binding pairs;methods of their making and using
US5236081A (en) 1992-01-31 1993-08-17 Shape Inc. Compact disc package
DE69315847T2 (en) 1992-08-21 1998-06-25 Genentech Inc METHOD FOR TREATING A FAULT BROUGHT BY LFA-1
US5263743A (en) 1992-09-25 1993-11-23 Pharmagraphics, Inc. Package label
ATE173630T1 (en) 1992-12-29 1998-12-15 Genentech Inc TREATMENT OF INFLAMMATORY BOWEL DISEASE WITH INTERFERON-GAMMA INHIBITORS
US5830675A (en) 1993-03-10 1998-11-03 Cedars-Sinai Medical Center Methods for selectively detecting perinuclear anti-neutrophil cytoplasmic antibody of ulcerative colitis, primary sclerosing cholangitis, or type 1 autoimmune hepatitis
DE69424700T2 (en) 1993-03-10 2000-11-09 Cedars Sinai Medical Center Lo Method for the selective detection of perinuclear anti-neutrophil cytoplasmic antibodies in ulcerative colitis or primary sclerotic cholangitis
GB9307503D0 (en) 1993-04-13 1993-06-02 Kodak Ltd Method of making a photographic developer solution
GB9325182D0 (en) 1993-12-08 1994-02-09 T Cell Sciences Inc Humanized antibodies or binding proteins thereof specific for t cell subpopulations exhibiting select beta chain variable regions
US5681699A (en) 1994-02-11 1997-10-28 Cedars-Sinai Medical Center Methods of diagnosing ulcerative colitis and Crohn's disease
DE69523586T2 (en) 1994-05-17 2002-07-18 Cedars Sinai Medical Ct Los An SCREENING METHOD FOR DISEASE CROHN BY USE OF TNF MICROSATELLITE ALLELS
US5494920A (en) 1994-08-22 1996-02-27 Eli Lilly And Company Methods of inhibiting viral replication
US5491063A (en) 1994-09-01 1996-02-13 Hoffmann-La Roche Inc. Methods for in-solution quenching of fluorescently labeled oligonucleotide probes
US5691151A (en) 1994-10-07 1997-11-25 Regents Of University Of California Methods of screening for ulcerative colitis and crohn's disease by detecting VH3-15 autoantibody and panca
US7597886B2 (en) 1994-11-07 2009-10-06 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US7820798B2 (en) 1994-11-07 2010-10-26 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
WO1996014328A1 (en) 1994-11-07 1996-05-17 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US20030198640A1 (en) 1994-11-07 2003-10-23 Human Genome Sciences, Inc. Methods and compositions for treating inflammatory bowel diseases relating to human tumor necrosis factor-gamma-beta
US6599719B2 (en) 1994-11-07 2003-07-29 Human Genome Sciences, Inc. Nucleic acid molecules encoding tumor necrosis factor-gamma-alpha
US20030129189A1 (en) 1994-11-07 2003-07-10 Guo-Liang Yu Tumor necrosis factor-gamma
US6824767B2 (en) 1994-11-07 2004-11-30 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
JPH08248832A (en) 1995-03-06 1996-09-27 Mita Ind Co Ltd Image forming device
US5518488A (en) 1995-03-20 1996-05-21 Schluger; Allen CD holder of cardboard and method of construction
US5607879A (en) 1995-06-28 1997-03-04 Taiwan Semiconductor Manufacturing Company Ltd. Method for forming buried plug contacts on semiconductor integrated circuits
US6696248B1 (en) 1995-08-18 2004-02-24 Morphosys Ag Protein/(poly)peptide libraries
CA2229043C (en) 1995-08-18 2016-06-07 Morphosys Gesellschaft Fur Proteinoptimierung Mbh Protein/(poly)peptide libraries
US6632976B1 (en) 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
US5840300A (en) 1995-09-11 1998-11-24 Trustees Of The University Of Pennsylvania Methods and compositions comprising single chain recombinant antibodies
US5942390A (en) 1996-01-12 1999-08-24 Cedars-Sinai Medical Center Method of diagnosing predisposition for ulcerative colitis in Jewish population by detection of interleukin-1 receptor antagonist polymorphism
US7357927B2 (en) 1996-03-12 2008-04-15 Human Genome Sciences, Inc. Death domain containing receptors
US6713061B1 (en) 1996-03-12 2004-03-30 Human Genome Sciences, Inc. Death domain containing receptors
US5590769A (en) 1996-03-20 1997-01-07 Lin; Shi-Ping Individual CD case
CA2250118C (en) 1996-03-26 2009-09-29 Michael S. Kopreski Method enabling use of extracellular rna extracted from plasma or serum to detect, monitor or evaluate cancer
US6025539A (en) 1996-04-09 2000-02-15 Mayo Foundation For Medical Education And Research IL-5 transgenic mouse
US5916748A (en) 1996-04-12 1999-06-29 Cedars-Sinai Medical Center Method of diagnosing a clinical subtype of crohn's disease with features of ulcerative colitis
US6074835A (en) 1996-04-12 2000-06-13 Regents Of The Univ. Of California Diagnosis, prevention and treatment of ulcerative colitis, and clinical subtypes thereof, using histone H1
US5874233A (en) 1996-04-12 1999-02-23 Cedars-Sinai Medical Center Methods of diagnosing a clinical subtype of Crohn's disease with features of ulcerative colitis
AU2438397A (en) 1996-04-12 1997-11-07 Cedars-Sinai Medical Center Methods of determining the risk of pouchitis development
US5683698A (en) 1996-08-02 1997-11-04 New England Deaconess Hospital Formulation for alleviating symptoms associated with arthritis
JPH1078528A (en) 1996-09-03 1998-03-24 Fujitsu Ltd Optical multiplexer/demultiplexer and wavelength division multiplexing module
US6034102A (en) 1996-11-15 2000-03-07 Pfizer Inc Atherosclerosis treatment
US6114395A (en) 1996-11-15 2000-09-05 Pfizer Inc. Method of treating atherosclerosis
US7514232B2 (en) 1996-12-06 2009-04-07 Becton, Dickinson And Company Method for detecting T cell response to specific antigens in whole blood
US20040013664A1 (en) 1997-01-14 2004-01-22 Gentz Reiner L. Tumor necrosis factor receptors 6 alpha & 6 beta
EP1007659B1 (en) 1997-01-14 2007-06-27 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6 alpha & 6 beta
US7285267B2 (en) 1997-01-14 2007-10-23 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6α & 6β
US6197928B1 (en) 1997-03-14 2001-03-06 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
US6183951B1 (en) 1997-04-11 2001-02-06 Prometheus Laboratories, Inc. Methods of diagnosing clinical subtypes of crohn's disease with characteristic responsiveness to anti-Th1 cytokine therapy
US5968741A (en) 1997-04-11 1999-10-19 Cedars-Sinai Medical Center Methods of diagnosing a medically resistant clinical subtype of ulcerative colitis
US20030129215A1 (en) 1998-09-24 2003-07-10 T-Ram, Inc. Medical devices containing rapamycin analogs
DK1034298T3 (en) 1997-12-05 2012-01-30 Scripps Research Inst Humanization of murine antibody
US6297367B1 (en) 1997-12-30 2001-10-02 Chiron Corporation Polynucleotide encoding TNFL1
US20020006613A1 (en) 1998-01-20 2002-01-17 Shyjan Andrew W. Methods and compositions for the identification and assessment of cancer therapies
US6607879B1 (en) 1998-02-09 2003-08-19 Incyte Corporation Compositions for the detection of blood cell and immunological response gene expression
EP1071458A4 (en) 1998-03-13 2005-02-16 Dana Farber Cancer Inst Inc Humanized antibody and uses thereof
US5947281A (en) 1998-07-06 1999-09-07 Kaneff; Mitchell S. Unfolding disc holder
US20050123938A1 (en) 1999-01-06 2005-06-09 Chondrogene Limited Method for the detection of osteoarthritis related gene transcripts in blood
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7368527B2 (en) 1999-03-12 2008-05-06 Human Genome Sciences, Inc. HADDE71 polypeptides
CA2267481A1 (en) 1999-03-30 2000-09-30 Gabriel Pulido-Cejudo Critical interdependency: from the role of estrogen on breast cancer to the susceptibility of women towards hiv infection
AU5124500A (en) 1999-04-30 2000-11-17 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US6539374B2 (en) 1999-06-03 2003-03-25 Microsoft Corporation Methods, apparatus and data structures for providing a uniform representation of various types of information
KR20020016833A (en) 1999-06-15 2002-03-06 뉴트리-로직스, 인크. Nutrient Formulations for Disease Reduction, and Related Treatment and Component Screening Methods
US6692916B2 (en) 1999-06-28 2004-02-17 Source Precision Medicine, Inc. Systems and methods for characterizing a biological condition or agent using precision gene expression profiles
ES2353268T3 (en) 1999-07-02 2011-02-28 Morphosys Ag GENERATION OF SPECIFIC UNION ELEMENTS THAT JOIN (POLI) PEPTIDES CODIFIED BY FRAGMENTS OF GENOMIC DNA OR EST.
US20030204075A9 (en) 1999-08-09 2003-10-30 The Snp Consortium Identification and mapping of single nucleotide polymorphisms in the human genome
US6376176B1 (en) 1999-09-13 2002-04-23 Cedars-Sinai Medical Center Methods of using a major histocompatibility complex class III haplotype to diagnose Crohn's disease
US6762042B2 (en) 1999-12-09 2004-07-13 Merck & Co., Inc. DNA molecules encoding human NHL a DNA helicase
EP1276899A2 (en) 1999-12-10 2003-01-22 Whitehead Institute For Biomedical Research Ibd-related polymorphisms
US6869762B1 (en) 1999-12-10 2005-03-22 Whitehead Institute For Biomedical Research Crohn's disease-related polymorphisms
WO2001055321A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
US20020165137A1 (en) 2000-01-31 2002-11-07 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU2001245371A1 (en) 2000-02-28 2001-09-12 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulators of type-1 tumor necrosis factor receptor and other cytokine receptor shedding
US6348316B1 (en) 2000-04-12 2002-02-19 Cedars-Sinai Medical Center Genetic testing for determining the risk of pouchitis development
US20110131679A2 (en) 2000-04-19 2011-06-02 Thomas La Rosa Rice Nucleic Acid Molecules and Other Molecules Associated with Plants and Uses Thereof for Plant Improvement
EP1286664B1 (en) 2000-05-12 2007-07-25 Oregon Health and Science University Combination of low dose estrogen and immunotherapeutic agent for treating immune diseases
US6972187B2 (en) 2000-05-12 2005-12-06 Millennium Pharmaceuticals, Inc. OAT5 molecules and uses therefor
US7138237B1 (en) 2000-05-19 2006-11-21 Cedars-Sinai Medical Center Diagnosis, prevention and treatment of Crohn's disease using the OmpC antigen
US6571638B2 (en) 2000-06-30 2003-06-03 Sawtek, Inc. Surface-acoustic-wave pressure sensor and associated methods
WO2002004643A1 (en) 2000-07-07 2002-01-17 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
CN1310942C (en) 2000-08-04 2007-04-18 路德维希癌症研究院 Suppressor gene
US20020019837A1 (en) 2000-08-11 2002-02-14 Balnaves James A. Method for annotating statistics onto hypertext documents
US6824989B1 (en) 2000-09-01 2004-11-30 Upstate Biotechnology, Inc. Recombinant monoclonal antibody to phosphotyrosine-containing proteins
US20070037165A1 (en) 2000-09-08 2007-02-15 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US6812339B1 (en) 2000-09-08 2004-11-02 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US20020048566A1 (en) 2000-09-14 2002-04-25 El-Deiry Wafik S. Modulation of cellular apoptosis and methods for treating cancer
AU2126502A (en) 2000-10-03 2002-04-15 Gene Logic, Inc. Gene expression profiles in granulocytic cells
AU1092602A (en) 2000-10-24 2002-05-06 New Ind Res Organization Genomes participating in rheumatoid arthritis, method of the diagnosis thereof, method of evaluating the onset possibility thereof, diagnostic kit for detectingthem and therapeutic method and remedies for rheumatoid arthritis
USH2191H1 (en) 2000-10-24 2007-06-05 Snp Consortium Identification and mapping of single nucleotide polymorphisms in the human genome
WO2002044426A2 (en) 2000-10-30 2002-06-06 Regents Of The University Of Michigan Nod2 nucleic acids and proteins
US7820447B2 (en) 2000-12-22 2010-10-26 Sagres Discovery Inc. Compositions and methods for cancer
US7892730B2 (en) 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US20040072154A1 (en) 2000-12-22 2004-04-15 Morris David W. Novel compositions and methods for cancer
US20030092019A1 (en) 2001-01-09 2003-05-15 Millennium Pharmaceuticals, Inc. Methods and compositions for diagnosing and treating neuropsychiatric disorders such as schizophrenia
WO2003008583A2 (en) 2001-03-02 2003-01-30 Sagres Discovery Novel compositions and methods for cancer
US20020182274A1 (en) 2001-03-21 2002-12-05 Kung-Ming Lu Methods for inhibiting cancer growth, reducing infection and promoting general health with a fermented soy extract
US20050261219A1 (en) 2001-05-18 2005-11-24 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US20050182007A1 (en) 2001-05-18 2005-08-18 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20050143333A1 (en) 2001-05-18 2005-06-30 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US6905827B2 (en) 2001-06-08 2005-06-14 Expression Diagnostics, Inc. Methods and compositions for diagnosing or monitoring auto immune and chronic inflammatory diseases
CA2391711A1 (en) 2001-06-26 2002-12-26 University Of Guelph Non-radioactive assay of lipopolysaccharide kinases
US20070082337A1 (en) 2004-01-27 2007-04-12 Compugen Ltd. Methods of identifying putative gene products by interspecies sequence comparison and biomolecular sequences uncovered thereby
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
WO2003025148A2 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
GB0124315D0 (en) 2001-10-10 2001-11-28 Oxagen Ltd Inflammatory bowel disease
KR20040064275A (en) 2001-11-09 2004-07-16 소스 프리시전 메디슨, 인코포레이티드 Identification, monitoring and treatment of disease and characterization of biological condition using gene expression profiles
AU2002352799A1 (en) 2001-11-20 2003-06-10 Oncomedx, Inc. Methods for evaluating drug-resistance gene expression in the cancer patient
US7871619B2 (en) 2001-11-30 2011-01-18 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US20070237770A1 (en) 2001-11-30 2007-10-11 Albert Lai Novel compositions and methods in cancer
WO2003053220A2 (en) 2001-12-17 2003-07-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of inflammatory bowel disease
FI20020078A (en) 2002-01-15 2003-07-16 Danisco Stimulation of the immune system by polydextrosis
US6878518B2 (en) 2002-01-22 2005-04-12 The Trustees Of The University Of Pennsylvania Methods for determining steroid responsiveness
US20070015271A1 (en) 2002-04-04 2007-01-18 Rosen Craig A Human secreted proteins
US7745180B2 (en) 2002-04-24 2010-06-29 Hitachi Chemical Co., Ltd. Device and method for high-throughput quantification of mRNA from whole blood
WO2003099773A1 (en) 2002-05-24 2003-12-04 Millennium Pharmaceuticals, Inc. Ccr9 inhibitors and methods of use thereof
AU2003232872A1 (en) 2002-05-27 2003-12-12 Ostergotlands Lans Landsting Method for determining immune system affecting compounds
US20040053263A1 (en) 2002-08-30 2004-03-18 Abreu Maria T. Mutations in NOD2 are associated with fibrostenosing disease in patients with Crohn's disease
TW200409759A (en) 2002-09-25 2004-06-16 Wyeth Corp Substituted 4-(indazol-3-yl)phenols
WO2004035537A2 (en) 2002-10-16 2004-04-29 Euro-Celtique S.A. Antibodies that bind cell-associated ca 125/o772p and methods of use thereof
AU2003304386A1 (en) 2002-10-30 2005-02-25 The Center For Blood Research, Inc. Methods for treating and preventing apoptosis-related diseases using rna interfering agents
WO2004048600A2 (en) 2002-11-22 2004-06-10 Emory University Diagnostic tests and methods for diagnosing inflammatory bowel disease
CA2507080C (en) 2002-11-27 2015-01-06 Biogen Idec Ma Inc. Humanized antibodies against monocyte chemotactic proteins
ATE515514T1 (en) 2002-12-23 2011-07-15 Schering Corp USES OF MAMMAL CYTOKINE IL-23 ; RELATED REAGENTS
AU2002953533A0 (en) 2002-12-24 2003-01-16 Arthron Limited Fc receptor modulating compounds and compositions
US20070020637A1 (en) 2003-01-21 2007-01-25 Research Association For Biotechnology Full-length cDNA
US8071304B2 (en) 2003-04-05 2011-12-06 The Johns Hopkins University Methods for detecting a polymorphism in the NFKB1 gene promoter
US7662569B2 (en) 2003-04-11 2010-02-16 Cedars-Sinai Medical Center Methods of assessing Crohn's disease patient phenotype by I2 serologic response
US20060100132A1 (en) 2003-06-06 2006-05-11 Brit Corneliussen Method for diagnosing inflammatory bowel disease
CA2536086C (en) 2003-08-20 2013-03-19 University Of Miami Inhibition of dr3 in the treatment of allergic lung inflammation
WO2005018436A2 (en) 2003-08-26 2005-03-03 The Trustees Of Boston University Methods for the diagnosis, prognosis and treatment of metabolic syndrome
US20050163764A1 (en) 2003-09-22 2005-07-28 Yale University Treatment with agonists of toll-like receptors
US20070161003A1 (en) 2003-09-29 2007-07-12 Morris David W Novel therapeutic targets in cancer
US20050101889A1 (en) 2003-11-06 2005-05-12 Freeman Gary A. Using chest velocity to process physiological signals to remove chest compression artifacts
US20070054278A1 (en) 2003-11-18 2007-03-08 Applera Corporation Polymorphisms in nucleic acid molecules encoding human enzyme proteins, methods of detection and uses thereof
EP1692519B1 (en) 2003-12-03 2010-06-02 Glycominds Ltd. Method for diagnosing diseases based on levels of anti-glycan antibodies
JP2008504803A (en) 2004-01-09 2008-02-21 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Cell type-specific pattern of gene expression
US7691379B2 (en) 2004-04-12 2010-04-06 Medimmune, Llc Anti-IL-9 antibody formulations
WO2005114469A1 (en) 2004-04-19 2005-12-01 Kartoo Sa Method and device for searching and customisably storing results
US7759079B2 (en) 2004-05-13 2010-07-20 Prometheus Laboratories Inc. Methods of diagnosing inflammatory bowel disease
US20060154276A1 (en) 2004-05-13 2006-07-13 Prometheus Laboratories Inc. Methods of diagnosing inflammatory bowel disease
US7407753B2 (en) 2004-05-24 2008-08-05 Yissum Research Development Company Of The Hebrewuniversityofjerusalem Methods, kits and pharmaceutical compositions for diagnosing, delaying onset of, preventing and/or treating osteoporosis
CN101426930A (en) 2004-05-25 2009-05-06 日立化成工业株式会社 Method of measuring cancer susceptibility
CA2573293A1 (en) 2004-07-10 2006-02-16 Alexion Pharmaceuticals, Inc. Methods for discovering antibodies specific to cancer cells and antibodies discovered thereby
WO2006020430A2 (en) 2004-08-12 2006-02-23 Genentech, Inc. Novel composition and methods for the treatment of immune related disease
TWI309240B (en) 2004-09-17 2009-05-01 Hoffmann La Roche Anti-ox40l antibodies
AR051444A1 (en) 2004-09-24 2007-01-17 Centocor Inc PROTEINS DERIVED FROM IL-23P40 SPECIFIC IMMUNOGLOBULIN, COMPOSITIONS, EPITHOPES, METHODS AND USES
EP1805319B1 (en) 2004-09-29 2014-09-03 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Recombinant human t2 rnase and uses thereof
US20090297563A1 (en) 2004-10-27 2009-12-03 Anders Borglum Diagnosis And Treatment of Immune-Related Diseases
WO2006048291A2 (en) 2004-11-03 2006-05-11 Almac Diagnostics Limited Transcriptome microarray technology and methods of using the same
EP2270512B1 (en) 2004-12-08 2016-03-23 Cedars-Sinai Medical Center Methods for diagnosis of Crohn's disease
US7914990B2 (en) 2005-01-13 2011-03-29 Progenika Biopharma, S.A. Methods and products for in vitro genotyping
EP1875246B1 (en) 2005-04-15 2016-10-05 CanImGuide Therapeutics AB Diagnostic method for detecting cancer by measuring amount of a cytokine like il-6
ATE512234T1 (en) 2005-04-28 2011-06-15 Hitachi Chemical Res Ct Inc EX-VIVO GENE EXPRESSION IN WHOLE BLOOD AS A MODEL FOR ASSESSING INDIVIDUAL VARIATION AGAINST FOOD ADDITIVES
CN101198624B (en) 2005-05-06 2012-10-10 津莫吉尼蒂克斯公司 IL-31 monoclonal antibodies and methods of use
ES2432564T3 (en) 2005-05-10 2013-12-04 Biogen Idec Ma Inc. Treatment and evaluation of inflammatory disorders
ES2301280A1 (en) 2005-05-16 2008-06-16 Fina Biotech S.L.U. Method for the diagnosis of alzheimer's disease
WO2006127900A2 (en) 2005-05-25 2006-11-30 Biogen Idec Ma Inc. Tl1a in the treatment of disease
TW201444869A (en) 2005-06-30 2014-12-01 Abbvie Inc IL-12/P40 binding proteins
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
CN101253199B (en) 2005-08-30 2019-06-14 迈阿密大学 Agonist, antagonist and the immunotoxin of immunomodulating tumor necrosis factor receptor 25 (TNFR25)
WO2007025989A2 (en) 2005-09-02 2007-03-08 Innogenetics N.V. Nucleic acid variants in the toll like receptor genes associated with altered innate immunity
EP1945814A2 (en) 2005-09-27 2008-07-23 Source MDX Gene expression profiling for identification monitoring and treatment of rheumatoid arthritis
US8234129B2 (en) 2005-10-18 2012-07-31 Wellstat Vaccines, Llc Systems and methods for obtaining, storing, processing and utilizing immunologic and other information of individuals and populations
US20090258848A1 (en) 2005-12-06 2009-10-15 The Johns Hopkins University Biomarkers for inflammatory bowel disease
US20070161031A1 (en) 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
AU2007249713A1 (en) 2006-01-10 2007-11-22 Zymogenetics, Inc. Methods of treating pain and inflammation in neuronal tissue using IL-31Ra and OSMRb antagonists
US7790862B2 (en) 2006-06-13 2010-09-07 Zymogenetics, Inc. IL-17 and IL-23 antagonists and methods of using the same
US20100019016A1 (en) 2006-03-23 2010-01-28 Edoga John K Vascular Anastomotic Staplers
WO2007117611A2 (en) 2006-04-07 2007-10-18 Hitachi Chemical Co., Ltd. Enhanced t cell receptor-mediated tumor necrosis factor superfamily and chemokine mrna expression in peripheral blood leukocytes in patients with crohn's disease
CA2654761A1 (en) 2006-06-09 2007-12-13 The University Of British Columbia Interferon gamma polymorphisms as indicators of subject outcome in critically ill subjects
NZ573646A (en) 2006-06-12 2012-04-27 Wyeth Llc Single-chain multivalent binding proteins with effector function
EP2049691A4 (en) 2006-07-26 2010-06-16 Genizon Biosciences Inc Crohn disease susceptibility gene
US7993832B2 (en) 2006-08-14 2011-08-09 Xdx, Inc. Methods and compositions for diagnosing and monitoring the status of transplant rejection and immune disorders
US20080293582A1 (en) 2006-08-30 2008-11-27 Xilin Li Markers and Methods for Assessing and Treating Ulcerative Colitis and Related Disorders Using a 43 Gene Panel
EP2636754B1 (en) 2006-09-11 2015-03-04 Celera Corporation Genetic polymorphisms associated with psoriasis, methods of detection and uses thereof
CN101516839A (en) 2006-09-25 2009-08-26 贝林格尔.英格海姆国际有限公司 Compounds which modulate the cb2 receptor
WO2008048902A2 (en) 2006-10-13 2008-04-24 Cedars-Sinai Medical Center Methods of using single nucleotide polymorphisms in the il23r gene to predict or diagnose inflammatory bowel disease
US20100267575A1 (en) 2006-10-17 2010-10-21 Childrens Hospital Medical Center Gene array technique for predicting response in inflammatory bowel diseases
WO2008048984A2 (en) 2006-10-18 2008-04-24 St. Jude Children's Research Hospital Methods and compositions for preparing a universal influenza vaccine
US20100291551A1 (en) 2006-11-17 2010-11-18 Genizon Biosciences Inc. Genemap of the human associated with crohn's disease
US20080131887A1 (en) 2006-11-30 2008-06-05 Stephan Dietrich A Genetic Analysis Systems and Methods
EP2097538A4 (en) 2006-12-07 2011-11-30 Switchgear Genomics Transcriptional regulatory elements of biological pathways, tools, and methods
US20090186034A1 (en) 2006-12-19 2009-07-23 Genetech, Inc. Gene expression markers for inflammatory bowel disease
US7691980B2 (en) 2007-01-09 2010-04-06 Bio-Rad Laboratories, Inc. Enhanced capacity and purification of antibodies by mixed mode chromatography in the presence of aqueous-soluble nonionic organic polymers
US9896511B2 (en) 2007-01-10 2018-02-20 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies that bind to TL1A and methods of treating inflammatory or autoimmune disease comprising administering such antibodies
US20110217310A1 (en) 2007-01-10 2011-09-08 Siegel Richard M Blockade of tl1a-dr3 interactions to ameliorate t cell mediated disease pathology
US20100021917A1 (en) 2007-02-14 2010-01-28 Cedars-Sinai Medical Center Methods of using genes and genetic variants to predict or diagnose inflammatory bowel disease
US20100190162A1 (en) 2007-02-26 2010-07-29 Cedars-Sinai Medical Center Methods of using single nucleotide polymorphisms in the tl1a gene to predict or diagnose inflammatory bowel disease
WO2008106579A2 (en) 2007-02-28 2008-09-04 Cedars-Sinai Medical Center The role of il-12, il-23 and il-17 receptors in inflammatory bowel disease
WO2008137762A2 (en) 2007-05-04 2008-11-13 Cedars-Sinai Medical Center Methods of diagnosis and treatment of crohn's disease
WO2010039931A2 (en) 2008-10-01 2010-04-08 Cedars-Sinai Medical Center Methods of using il17rd and il23-il17 pathway genes to diagnose crohn's disease
US20150337378A1 (en) 2007-02-28 2015-11-26 Cedars-Sinai Medical Center Methods of diagnosis and treatment of inflammatory bowel disease
JP2010519895A (en) 2007-03-02 2010-06-10 ユニヴェルシト ド リエージュ Methods for determining genotypes at Crohn's disease locus
US20100015156A1 (en) 2007-03-06 2010-01-21 Cedars-Sinai Medical Center Diagnosis of inflammatory bowel disease in children
WO2008106987A1 (en) 2007-03-06 2008-09-12 Eesti Biokeskus A method to determine single nucleotide polymorphisms and mutations in nucleic acid sequence
WO2008112990A2 (en) 2007-03-15 2008-09-18 Cedars-Sinai Medical Center Methods of diagnosis and treatment of crohn's disease
US8486640B2 (en) 2007-03-21 2013-07-16 Cedars-Sinai Medical Center Ileal pouch-anal anastomosis (IPAA) factors in the treatment of inflammatory bowel disease
US20100184050A1 (en) 2007-04-26 2010-07-22 Cedars-Sinai Medical Center Diagnosis and treatment of inflammatory bowel disease in the puerto rican population
US8153443B2 (en) 2007-05-10 2012-04-10 Cedars-Sinai Medical Center Characterization of the CBir1 antigenic response for diagnosis and treatment of Crohn's disease
US9305137B1 (en) 2007-05-18 2016-04-05 Cedars-Sinai Medical Center Methods of identifying the genetic basis of a disease by a combinatorial genomics approach, biological pathway approach, and sequential approach
US20150086567A1 (en) * 2011-03-25 2015-03-26 Cedars-Sinai Medical Center Role of ifng methylation in inflammatory bowel disease
JP2010527937A (en) 2007-05-18 2010-08-19 メディミューン,エルエルシー IL-9 in fibrotic and inflammatory diseases
US20150376707A1 (en) 2007-05-18 2015-12-31 Cedars-Sinai Medical Center Methods of diagnosing and treating inflammatory bowel disease
WO2011088237A1 (en) 2010-01-13 2011-07-21 Cedars-Sinai Medical Center Methods of using znf365 genetic variants to diagnose crohn's disease
US20120190698A1 (en) 2011-01-25 2012-07-26 Cedars-Sinai Medical Center Methods of predicting thiopurine response
CN101821408A (en) 2007-08-09 2010-09-01 奥塔戈大学 Differentiate the individual method that is in thio-purine drug resistance and intolerance risk
AU2008304205B8 (en) 2007-09-26 2014-08-28 Navigenics, Inc. Methods and systems for genomic analysis using ancestral data
WO2009052512A2 (en) 2007-10-19 2009-04-23 Cedars-Sinai Medical Center Methods of using genetic variants to diagnose and predict inflammatory bowel disease
EP2220122A2 (en) 2007-11-13 2010-08-25 Teva Biopharmaceuticals USA, Inc. Humanized antibodies against tl1a
AU2008334095A1 (en) * 2007-11-29 2009-06-11 Genentech, Inc. Gene expression markers for inflammatory bowel disease
US7947451B2 (en) 2007-11-30 2011-05-24 Celera Corporation Genetic polymorphisms associated with psoriasis, methods of detection and uses thereof
CN101469270B (en) 2007-12-27 2012-08-22 牛斌 Industrial continuous plastic cracker
US7773519B2 (en) 2008-01-10 2010-08-10 Nuova Systems, Inc. Method and system to manage network traffic congestion
US8093364B2 (en) 2008-01-18 2012-01-10 Bio-Rad Laboratories, Inc. Enhanced purification of antibodies and antibody fragments by apatite chromatography
CA2714713C (en) 2008-02-19 2018-02-27 The Children's Hospital Of Philadelphia Identification of pediatric onset inflammatory bowel disease loci and methods of use thereof for the diagnosis and treatment of the same
US20090307180A1 (en) 2008-03-19 2009-12-10 Brandon Colby Genetic analysis
WO2009143278A2 (en) 2008-05-20 2009-11-26 Cedars-Sinai Medical Center Methods of diagnosing and characterizing cannabinoid signaling in crohn's disease
US20110111418A1 (en) 2008-06-23 2011-05-12 Cedars-Sinai Medical Center Use of iron-related pathways and genes for treatment and diagnosis of parkinson's disease
US20100008934A1 (en) 2008-07-02 2010-01-14 Celera Corporation Genetic polymorphisms associated with autoinflammatory diseases, methods of detection and uses thereof
CN102171365B (en) 2008-08-25 2014-01-29 森托科尔奥索生物科技公司 Biomarkers for anti-TNF treatment in ulcerative colitis and related disorders
US20110136113A1 (en) 2008-08-27 2011-06-09 Sysmex Corporation Marker for detecting il-17-producing helper t cell, and method for detecting il-17-producing helper t cell
CN102197051A (en) 2008-08-28 2011-09-21 惠氏有限责任公司 Uses of IL-22, IL-17, and IL-1 family cytokines in autoimmune diseases
JP2010088432A (en) 2008-09-09 2010-04-22 Kansetsu Saisei Kenkyusho:Kk Polymorphism relating to effectiveness-side effect expression by tnf inhibitor therapy and use thereof
WO2010034443A1 (en) 2008-09-29 2010-04-01 F. Hoffmann-La Roche Ag Antibodies against human il 17 and uses thereof
CA2740205A1 (en) 2008-10-21 2010-04-22 Morehouse School Of Medicine Methods for haplotype determination by haplodissection
US20110189685A1 (en) 2008-10-22 2011-08-04 Cedars-Sinai Medical Center Methods of using jak3 genetic variants to diagnose and predict crohn's disease
WO2010049933A1 (en) 2008-10-30 2010-05-06 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Efficient expression of truncated human rnaset2 in e. coli
AU2009314259B2 (en) 2008-11-11 2015-06-11 Nestec S.A. Methods for prediction of inflammatory bowel disease (IBD) using serologic markers
US20110229471A1 (en) 2008-11-26 2011-09-22 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-tnf alpha therapy in inflammatory bowel disease
US20170044615A1 (en) 2008-12-24 2017-02-16 Cedars-Sinai Medical Center METHODS OF PREDICTING MEDICALLY REFRACTIVE ULCERATIVE COLITIS (mrUC) REQUIRING COLECTOMY
WO2010075579A2 (en) 2008-12-24 2010-07-01 Cedars-Sinai Medical Center Methods of predicting medically refractive ulcerative colitis (mr-uc) requiring colectomy
WO2010075584A1 (en) 2008-12-24 2010-07-01 Cedars-Sinai Medical Center Methods of diagnosing and predicting crohns disease from childhood hygiene and serological profiles
US20120041082A1 (en) 2009-01-13 2012-02-16 Cedars-Sinai Medical Center Methods of using smad3 and jak2 genetic variants to diagnose and predict inflammatory bowel disease
AU2010228990A1 (en) 2009-03-24 2011-10-27 Teva Biopharmaceuticals Usa, Inc. Humanized antibodies against LIGHT and uses thereof
US20120073585A1 (en) 2009-04-08 2012-03-29 Cedars-Sinai Medical Center Methods of predicting complication and surgery in crohn's disease
EP2419529B1 (en) 2009-04-14 2015-05-20 Nestec S.A. Inflammatory bowel disease prognostics
JP5822822B2 (en) 2009-04-17 2015-11-24 ニューヨーク ユニバーシティ Peptides that target TNF family receptors and antagonize TNF action, compositions, methods and uses thereof
JP2011004390A (en) 2009-05-18 2011-01-06 Canon Inc Imaging device, imaging system, and method for driving imaging device
AU2010259022B2 (en) 2009-06-08 2016-05-12 Singulex, Inc. Highly sensitive biomarker panels
JP6073133B2 (en) 2009-06-24 2017-02-01 クルナ・インコーポレーテッド Treatment of TNFR2-related diseases by suppression of natural antisense transcripts against tumor necrosis factor receptor 2 (TNFR2)
US20110033486A1 (en) 2009-07-20 2011-02-10 Abbas Alexander R Gene expression markers for crohn's disease
WO2011017120A1 (en) 2009-07-27 2011-02-10 Cedars-Sinai Medical Center Use of ccr9, ccl25, batf and il17/il23 pathway variants to diagnose and treat inflammatory bowel disease
ES2586837T3 (en) 2009-08-03 2016-10-19 University Of Miami Method for in vivo expansion of regulatory T lymphocytes
US20130136720A1 (en) 2010-01-15 2013-05-30 Cedars-Sinai Medical Center Methods of using fut2 genetic variants to diagnose crohn's disease
WO2011088306A1 (en) 2010-01-15 2011-07-21 Cedars-Sinai Medical Center Methods of using genetic variants to diagnose crohn's disease
WO2011106707A2 (en) 2010-02-26 2011-09-01 Human Genome Sciences, Inc. Antibodies that specifically bind to dr3
WO2011116111A1 (en) 2010-03-16 2011-09-22 Cedars-Sinai Medical Center Methods of using prdm1 genetic variants to prognose, diagnose and treat inflammatory bowel disease
WO2012025536A1 (en) 2010-08-25 2012-03-01 F. Hoffmann-La Roche Ag Antibodies against il-18r1 and uses thereof
US8859739B2 (en) 2010-09-16 2014-10-14 Baliopharm Ag Anti-huTNFR1 antibody and methods of use thereof for treatment
US8766034B2 (en) 2010-09-22 2014-07-01 Cedars-Sinai Medical Center TL1A model of inflammation fibrosis and autoimmunity
EP2630495B1 (en) 2010-10-18 2017-02-08 Nestec S.A. Methods for determining anti-drug antibody isotypes
JO3375B1 (en) 2010-11-08 2019-03-13 Regeneron Pharma Human antibodies to human tnf-like ligand 1a (tl1a)
WO2012071436A1 (en) 2010-11-24 2012-05-31 Genentech, Inc. Method of treating autoimmune inflammatory disorders using il-23r loss-of-function mutants
US9902996B2 (en) * 2011-02-11 2018-02-27 Cedars-Sinai Medical Center Methods of predicting the need for surgery in crohn's disease
KR20140008397A (en) 2011-02-17 2014-01-21 네스텍 소시에테아노님 Assays for detecting autoantibodies to anti-tnfa drugs
EP2689034B1 (en) 2011-03-25 2016-03-23 Cedars-Sinai Medical Center Role of ifng methylation in inflammatory bowel disease
CA2830351A1 (en) 2011-03-25 2012-10-04 Cedars-Sinai Medical Center Methods of diagnosing ulcerative colitis and crohn's disease
EP2689036B1 (en) 2011-03-25 2017-12-20 Cedars-Sinai Medical Center Methods of diagnosing and treating intestinal granulomas and low bone density in inflammatory bowel disease
CA2839792A1 (en) 2011-05-10 2012-11-15 Nestec S.A. Methods of disease activity profiling for personalized therapy management
KR20140104344A (en) 2011-05-20 2014-08-28 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 시크리터리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비시스 Blockade of tl1a-dr3 interactions to ameliorate t cell mediated disease pathology and antibodies thereof
BR112013030958B1 (en) 2011-06-03 2022-02-08 Xoma Technology Ltd ANTIBODY BINDING GROWTH TRANSFORMATION FACTOR BETA, PHARMACEUTICAL COMPOSITION, USES THEREOF, NUCLEIC ACID MOLECULE, EXPRESSION VECTOR, AND METHOD FOR PRODUCTION OF AN ANTIBODY
EP2720719A4 (en) 2011-06-15 2015-12-09 Glaxosmithkline Ip No 2 Ltd Method of selecting therapeutic indications
CN202109170U (en) 2011-06-21 2012-01-11 徐毓艺 Multi-seat rotary connecting device
US8478890B2 (en) 2011-07-15 2013-07-02 Damaka, Inc. System and method for reliable virtual bi-directional data stream communications with single socket point-to-multipoint capability
WO2013033627A2 (en) 2011-09-01 2013-03-07 The Regents Of The University Of California Diagnosis and treatment of arthritis using epigenetics
EP2565277A1 (en) 2011-09-05 2013-03-06 Progenika Biopharma, S.A. Method for predicting radiographic severity in ankylosing spondylitis
EA035018B1 (en) 2011-09-30 2020-04-17 Тева Фармасьютикал Австралия Пти Лтд. ANTIBODIES AGAINST TL1a AND USES THEREOF
SG11201401536QA (en) 2011-10-21 2014-05-29 Nestec Sa Methods for improving inflammatory bowel disease diagnosis
US20130123117A1 (en) 2011-11-16 2013-05-16 The Board Of Trustees Of The Leland Stanford Junior University Capture probe and assay for analysis of fragmented nucleic acids
KR20140097336A (en) 2011-11-23 2014-08-06 암젠 인크 Methods of treatment using an antibody against interferon gamma
US10233495B2 (en) 2012-09-27 2019-03-19 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
CN105102067B (en) 2013-01-02 2020-03-03 艾科诺斯科技股份有限公司 Antibodies that bind TL1A and uses thereof
CN103149371B (en) 2013-02-26 2015-12-23 哈药慈航制药股份有限公司 The application of biomarker in the feedback response medicine of preparation prediction 5-aminosalicylic acid treatment ulcerative colitis
EP2970398B1 (en) 2013-03-13 2024-05-08 The United States of America, as Represented by The Secretary, Department of Health and Human Services Prefusion rsv f proteins and their use
KR20210157418A (en) 2013-03-27 2021-12-28 세다르스-신나이 메디칼 센터 Mitigation and reversal of fibrosis and inflammation by inhibition of tl1a function and related signaling pathways
EP2996717A4 (en) 2013-05-17 2016-11-23 Cedars Sinai Medical Center Distinct effects of ifn-gamma and il-17 on tl1a modulated inflammation and fibrosis
ES2894963T3 (en) 2013-05-17 2022-02-16 Cedars Sinai Medical Center TNFSF15 and DcR3 variants associated with Crohn's disease
EP4105236A1 (en) 2013-07-19 2022-12-21 Cedars-Sinai Medical Center Anti-tl1a (tnfsf15) antibody for treatment of inflammatory bowel disease
MX2016002879A (en) * 2013-09-06 2016-08-17 Cedars Sinai Medical Center Systems, devices and methods for anti-tl1a therapy.
SG11201601844TA (en) 2013-09-13 2016-04-28 Beigene Ltd Anti-pd1 antibodies and their use as therapeutics and diagnostics
SG10201810298VA (en) 2013-11-13 2018-12-28 Pfizer Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
CA2947291A1 (en) * 2014-05-02 2015-11-05 Teva Pharmaceutical Industries Ltd. Treatment of crohn's disease with delayed-release 6-mercaptopurine
CA2946317A1 (en) 2014-05-02 2015-11-05 Cedars-Sinai Medical Center Methods of predicting medically refractive ulcerative colitis (mruc) requiring colectomy
WO2015191780A2 (en) 2014-06-10 2015-12-17 The General Hospital Corporation Ccctc-binding factor (ctcf) rna interactome
HUE049982T2 (en) 2014-11-14 2020-11-30 Hoffmann La Roche Antigen binding molecules comprising a tnf family ligand trimer
US20180051078A1 (en) 2015-03-16 2018-02-22 Cedars-Sinai Medical Center Methods of treating inflammatory bowel disease with ifn-gamma therapy
EP3286318A2 (en) 2015-04-22 2018-02-28 Mina Therapeutics Limited Sarna compositions and methods of use
KR20180004818A (en) 2015-05-15 2018-01-12 세다르스-신나이 메디칼 센터 Models, methods and compositions for the treatment of inflammatory bowel disease
TWI703158B (en) 2015-09-18 2020-09-01 美商希佛隆公司 Antibodies that specifically bind to tl1a
WO2017077715A1 (en) 2015-11-02 2017-05-11 La Jolla Institute For Allergy & Immunology Method and medicament for treating airway and/or lung diseases
CR20180365A (en) 2015-12-16 2018-09-28 Amgen Inc PROTEINS OF UNION TO THE ANTI-TL1A / ANTI-TNF-a BISPECTIVE ANTIGEN AND ITS USES
EP3430172A4 (en) 2016-03-17 2019-08-21 Cedars-Sinai Medical Center Methods of diagnosing inflammatory bowel disease through rnaset2
SG11201809793UA (en) 2016-05-09 2018-12-28 Bristol Myers Squibb Co Tl1a antibodies and uses thereof
KR20190016972A (en) 2016-05-20 2019-02-19 세다르스-신나이 메디칼 센터 Diagnosis of Inflammatory Bowel Disease Based on Gene
US10688412B2 (en) 2016-07-25 2020-06-23 Cehpalon, Inc. Affinity chromatography wash buffer
SG11201903737PA (en) 2016-10-26 2019-05-30 Cedars Sinai Medical Center Neutralizing anti-tl1a monoclonal antibodies
WO2018195328A1 (en) 2017-04-20 2018-10-25 Cedars-Sinai Medical Center Methods of predicting non-response to anti-tnf treatment in subjects with inflammatory bowel disease
WO2020056036A1 (en) 2018-09-12 2020-03-19 Strontium Neurogenics Inc. Compositions for treating gastroesophageal reflux and barrett's esophagus
CA3121162A1 (en) 2018-11-29 2020-06-04 Cedars-Sinai Medical Center Rnaset2 compositions and methods of treatment therewith
KR20220088529A (en) 2019-05-14 2022-06-27 프로메테우스 바이오사이언시즈, 인크. TL1A Patient Selection Methods, Systems and Devices

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11236393B2 (en) 2008-11-26 2022-02-01 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-TNFα therapy in inflammatory bowel disease
US11312768B2 (en) 2013-07-19 2022-04-26 Cedars-Sinai Medical Center Signature of TL1A (TNFSF15) signaling pathway
WO2021247770A1 (en) * 2020-06-03 2021-12-09 Cedars-Sinai Medical Center Methods and systems for measuring post-operative disease recurrence
WO2022140283A1 (en) * 2020-12-21 2022-06-30 Cedars-Sinai Medical Center Tl1a therapy compositions and methods of treatment therewith

Also Published As

Publication number Publication date
EP3430172A1 (en) 2019-01-23
US20220162703A1 (en) 2022-05-26
JP7082945B2 (en) 2022-06-09
US11186872B2 (en) 2021-11-30
KR102464372B1 (en) 2022-11-04
KR20220011802A (en) 2022-01-28
JP2022095664A (en) 2022-06-28
WO2017161342A1 (en) 2017-09-21
CN109462996A (en) 2019-03-12
KR20220153109A (en) 2022-11-17
JP2019509739A (en) 2019-04-11
KR20180127416A (en) 2018-11-28
EP3430172A4 (en) 2019-08-21

Similar Documents

Publication Publication Date Title
US20220162703A1 (en) Methods of diagnosing inflammatory bowel disease through rnaset2
AU2017293417B2 (en) Biomarkers for inflammatory bowel disease
EP2689036B1 (en) Methods of diagnosing and treating intestinal granulomas and low bone density in inflammatory bowel disease
US20100240043A1 (en) Methods of using genetic variants to diagnose and predict inflammatory bowel disease
US20150315643A1 (en) Blood transcriptional signatures of active pulmonary tuberculosis and sarcoidosis
US8153443B2 (en) Characterization of the CBir1 antigenic response for diagnosis and treatment of Crohn&#39;s disease
US20100184050A1 (en) Diagnosis and treatment of inflammatory bowel disease in the puerto rican population
Cortese et al. Correlative gene expression and DNA methylation profiling in lung development nominate new biomarkers in lung cancer
AU2009215441B2 (en) Identification of pediatric onset inflammatory bowel disease loci and methods of use thereof for the diagnosis and treatment of the same
Borghese et al. Identification of susceptibility genes for peritoneal, ovarian, and deep infiltrating endometriosis using a pooled sample‐based genome‐wide association study
WO2012022634A1 (en) Classification, diagnosis and prognosis of multiple myeloma
KR20230003560A (en) Methods for early detection of colorectal cancer, prediction of treatment response and prognosis
US20180208988A1 (en) Methods of diagnosis and treatment of inflammatory bowel disease
US10174380B2 (en) Methods for predicting multiple myeloma treatment response
JP5427352B2 (en) A method for determining the risk of developing obesity based on genetic polymorphisms associated with human body fat mass
WO2014072086A1 (en) Biomarkers for prognosis of lung cancer
US10428384B2 (en) Biomarkers for post-traumatic stress states
WO2017087735A1 (en) Method for treating crohn&#39;s disease
KR20240095481A (en) Methods of diagnosing inflammatory bowel disease through rnaset2
WO2010141362A1 (en) Compositions and methods for diagnosing the occurrence or likelihood of occurrence of testicular germ cell cancer
WO2015037681A1 (en) Test method for evaluating the risk of anti-thyroid drug-induced agranulocytosis, and evaluation kit
WO2010033825A2 (en) Genetic variants associated with abdominal aortic aneurysms
WO2017032865A1 (en) Method for evaluating individual radiosensitivity and the risk of adverse effects
KR20120001916A (en) Snp gene set for diagnosis of aspirin-induced asthma
JP2009207448A (en) Method for examining autoimmune disease, and method for screening prophylactic or curative agent for autoimmune disease

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: CEDARS-SINAI MEDICAL CENTER, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GONSKY, REBECCA;TARGAN, STEPHEN R.;DEEM, RICHARD L.;AND OTHERS;SIGNING DATES FROM 20181015 TO 20181105;REEL/FRAME:047434/0985

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT RECEIVED

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE